US20120071535A1 - Estrogen receptor modulators and uses thereof - Google Patents

Estrogen receptor modulators and uses thereof Download PDF

Info

Publication number
US20120071535A1
US20120071535A1 US13/234,035 US201113234035A US2012071535A1 US 20120071535 A1 US20120071535 A1 US 20120071535A1 US 201113234035 A US201113234035 A US 201113234035A US 2012071535 A1 US2012071535 A1 US 2012071535A1
Authority
US
United States
Prior art keywords
phenyl
indazol
acrylic acid
chloro
fluoro
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
US13/234,035
Other versions
US8299112B2 (en
Inventor
Nicholas D. Smith
Mehmet Kahraman
Steven P. Govek
Johnny Y. Nagasawa
Andiliy G. Lai
Jackaline D. Julien
Celine Bonnefous
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Seragon Pharmaceuticals Inc
Original Assignee
Aragon Pharmaceuticals Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=46599311&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20120071535(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Aragon Pharmaceuticals Inc filed Critical Aragon Pharmaceuticals Inc
Priority to US13/234,035 priority Critical patent/US8299112B2/en
Assigned to ARAGON PHARMACEUTICALS, INC. reassignment ARAGON PHARMACEUTICALS, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: BONNEFOUS, CELINE, JULIEN, JACKALINE D., GOVEK, STEVEN P., KAHRAMAN, MEHMET, LAI, ANDILIY G., NAGASAWA, JOHNNY Y., SMITH, NICHOLAS D.
Publication of US20120071535A1 publication Critical patent/US20120071535A1/en
Priority to US13/613,467 priority patent/US8455534B2/en
Application granted granted Critical
Publication of US8299112B2 publication Critical patent/US8299112B2/en
Assigned to SERAGON PHARMACEUTICALS, INC. reassignment SERAGON PHARMACEUTICALS, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: ARAGON PHARMACEUTICALS, INC.
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D231/00Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings
    • C07D231/54Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings condensed with carbocyclic rings or ring systems
    • C07D231/56Benzopyrazoles; Hydrogenated benzopyrazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/4151,2-Diazoles
    • A61K31/4161,2-Diazoles condensed with carbocyclic ring systems, e.g. indazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • A61K31/7052Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides
    • A61K31/7056Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing five-membered rings with nitrogen as a ring hetero atom
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • A61K31/7052Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides
    • A61K31/706Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom
    • A61K31/7064Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines
    • A61K31/7068Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines having oxo groups directly attached to the pyrimidine ring, e.g. cytidine, cytidylic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • A61K31/7052Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides
    • A61K31/706Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom
    • A61K31/7064Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines
    • A61K31/7068Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines having oxo groups directly attached to the pyrimidine ring, e.g. cytidine, cytidylic acid
    • A61K31/7072Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines having oxo groups directly attached to the pyrimidine ring, e.g. cytidine, cytidylic acid having two oxo groups directly attached to the pyrimidine ring, e.g. uridine, uridylic acid, thymidine, zidovudine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • A61K31/7052Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides
    • A61K31/706Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom
    • A61K31/7064Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines
    • A61K31/7076Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines containing purines, e.g. adenosine, adenylic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • A61K31/7052Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides
    • A61K31/706Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom
    • A61K31/7064Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines
    • A61K31/7076Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines containing purines, e.g. adenosine, adenylic acid
    • A61K31/708Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines containing purines, e.g. adenosine, adenylic acid having oxo groups directly attached to the purine ring system, e.g. guanosine, guanylic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • A61K31/7105Natural ribonucleic acids, i.e. containing only riboses attached to adenine, guanine, cytosine or uracil and having 3'-5' phosphodiester links
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/08Drugs for disorders of the alimentary tract or the digestive system for nausea, cinetosis or vertigo; Antiemetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/14Prodigestives, e.g. acids, enzymes, appetite stimulants, antidyspeptics, tonics, antiflatulents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/02Drugs for disorders of the urinary system of urine or of the urinary tract, e.g. urine acidifiers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • A61P15/08Drugs for genital or sexual disorders; Contraceptives for gonadal disorders or for enhancing fertility, e.g. inducers of ovulation or of spermatogenesis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • A61P19/10Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease for osteoporosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/06Antimigraine agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • A61P25/16Anti-Parkinson drugs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/18Antipsychotics, i.e. neuroleptics; Drugs for mania or schizophrenia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/24Antidepressants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/30Drugs for disorders of the nervous system for treating abuse or dependence
    • A61P25/32Alcohol-abuse
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/04Anorexiants; Antiobesity agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/20Antivirals for DNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/02Antithrombotic agents; Anticoagulants; Platelet aggregation inhibitors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/12Antihypertensives
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C215/00Compounds containing amino and hydroxy groups bound to the same carbon skeleton
    • C07C215/02Compounds containing amino and hydroxy groups bound to the same carbon skeleton having hydroxy groups and amino groups bound to acyclic carbon atoms of the same carbon skeleton
    • C07C215/04Compounds containing amino and hydroxy groups bound to the same carbon skeleton having hydroxy groups and amino groups bound to acyclic carbon atoms of the same carbon skeleton the carbon skeleton being saturated
    • C07C215/06Compounds containing amino and hydroxy groups bound to the same carbon skeleton having hydroxy groups and amino groups bound to acyclic carbon atoms of the same carbon skeleton the carbon skeleton being saturated and acyclic
    • C07C215/10Compounds containing amino and hydroxy groups bound to the same carbon skeleton having hydroxy groups and amino groups bound to acyclic carbon atoms of the same carbon skeleton the carbon skeleton being saturated and acyclic with one amino group and at least two hydroxy groups bound to the carbon skeleton
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D209/00Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D209/02Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom condensed with one carbocyclic ring
    • C07D209/04Indoles; Hydrogenated indoles
    • C07D209/08Indoles; Hydrogenated indoles with only hydrogen atoms or radicals containing only hydrogen and carbon atoms, directly attached to carbon atoms of the hetero ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D215/00Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems
    • C07D215/02Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom
    • C07D215/16Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D215/20Oxygen atoms
    • C07D215/22Oxygen atoms attached in position 2 or 4
    • C07D215/227Oxygen atoms attached in position 2 or 4 only one oxygen atom which is attached in position 2
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D217/00Heterocyclic compounds containing isoquinoline or hydrogenated isoquinoline ring systems
    • C07D217/22Heterocyclic compounds containing isoquinoline or hydrogenated isoquinoline ring systems with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to carbon atoms of the nitrogen-containing ring
    • C07D217/24Oxygen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D249/00Heterocyclic compounds containing five-membered rings having three nitrogen atoms as the only ring hetero atoms
    • C07D249/16Heterocyclic compounds containing five-membered rings having three nitrogen atoms as the only ring hetero atoms condensed with carbocyclic rings or ring systems
    • C07D249/18Benzotriazoles
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D265/00Heterocyclic compounds containing six-membered rings having one nitrogen atom and one oxygen atom as the only ring hetero atoms
    • C07D265/041,3-Oxazines; Hydrogenated 1,3-oxazines
    • C07D265/121,3-Oxazines; Hydrogenated 1,3-oxazines condensed with carbocyclic rings or ring systems
    • C07D265/141,3-Oxazines; Hydrogenated 1,3-oxazines condensed with carbocyclic rings or ring systems condensed with one six-membered ring
    • C07D265/181,3-Oxazines; Hydrogenated 1,3-oxazines condensed with carbocyclic rings or ring systems condensed with one six-membered ring with hetero atoms directly attached in position 2
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D277/00Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings
    • C07D277/60Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings condensed with carbocyclic rings or ring systems
    • C07D277/62Benzothiazoles
    • C07D277/68Benzothiazoles with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached in position 2
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D279/00Heterocyclic compounds containing six-membered rings having one nitrogen atom and one sulfur atom as the only ring hetero atoms
    • C07D279/101,4-Thiazines; Hydrogenated 1,4-thiazines
    • C07D279/141,4-Thiazines; Hydrogenated 1,4-thiazines condensed with carbocyclic rings or ring systems
    • C07D279/161,4-Thiazines; Hydrogenated 1,4-thiazines condensed with carbocyclic rings or ring systems condensed with one six-membered ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/10Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a carbon chain containing aromatic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/10Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a carbon chain containing aromatic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D409/00Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms
    • C07D409/02Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings
    • C07D409/10Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings linked by a carbon chain containing aromatic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/10Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings linked by a carbon chain containing aromatic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
    • C07D417/10Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings linked by a carbon chain containing aromatic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D495/00Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms
    • C07D495/02Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D495/04Ortho-condensed systems

Definitions

  • Described herein are compounds, including pharmaceutically acceptable salts, solvates, metabolites, prodrugs thereof, methods of making such compounds, pharmaceutical compositions comprising such compounds, and methods of using such compounds to treat, prevent or diagnose diseases or conditions that are estrogen sensitive, estrogen receptor dependent or estrogen receptor mediated.
  • the estrogen receptor (“ER”) is a ligand-activated transcriptional regulatory protein that mediates induction of a variety of biological effects through its interaction with endogenous estrogens. Endogenous estrogens include 17 ⁇ -estradiol and estrones. ER has been found to have two isoforms, ER- ⁇ and ER- ⁇ . Estrogens and estrogen receptors are implicated in a number of diseases or conditions, such as breast cancer, ovarian cancer, colon cancer, prostate cancer, endometrial cancer, uterine cancer, as well as others diseases or conditions.
  • compounds of Formula (I), or a pharmaceutically acceptable salt, or N-oxide thereof that diminish the effects of estrogens with estrogen receptors and/or lower the concentrations of estrogen receptors, and therefore, are useful as agents for the treatment or prevention of diseases or conditions in which the actions of estrogens and/or estrogen receptors are involved in the etiology or pathology of the disease or condition or contribute to at least one symptom of the disease or condition and wherein such actions of estrogens and/or estrogen receptors are undesirable.
  • compounds disclosed herein are estrogen receptor degrader compounds.
  • the compound of Formula (I), or a pharmaceutically acceptable salt, or N-oxide thereof is useful for the treatment of ER-related diseases or conditions including, but not limited to, ER-a dysfunction associated with cancer (e.g. bone cancer, breast cancer, colorectal cancer, endometrial cancer, prostate cancer, ovarian and uterine cancer), leiomyoma (e.g. uterine leiomyoma), central nervous system (CNS) defects (e.g. alcoholism, migraine), cardiovascular system defects (e.g. aortic aneurysm, susceptibility to myocardial infarction, aortic valve sclerosis, cardiovascular disease, coronary artery disease, hypertension), hematological system defects (e.g.
  • cancer e.g. bone cancer, breast cancer, colorectal cancer, endometrial cancer, prostate cancer, ovarian and uterine cancer
  • leiomyoma e.g. uterine leiomyoma
  • CNS central nervous system
  • deep vein thrombosis immune and inflammation diseases
  • susceptibility to infection e.g. hepatitis B, chronic liver disease
  • metabolic defects e.g. bone density, cholestasis, hypospadias, obesity, osteoarthritis, osteopenia, osteoporosis
  • neurological defects e.g. Alzheimer's disease, Parkinson's disease, migraine, vertigo
  • psychiatric defects e.g. anorexia nervosa, attention deficity hyperactivity disorder (ADHD), dementia, major depressive disorder, psychosis
  • reproductive defects e.g. age of menarche, endometriosis, infertility.
  • compounds of Formula (I) are estrogen receptor modulators.
  • the compound of Formula (I) is an estrogen receptor antagonist.
  • the compound of Formula (I) is an estrogen receptor degrader.
  • the compound of Formula (I) is an estrogen receptor antagonist as well as an estrogen receptor degrader.
  • the compound of Formula (I) displays minimal or no estrogen receptor agonist activity.
  • the compound of Formula (I) offers improved therapeutic activity characterized by complete or longer-lasting tumor regression, a lower incidence or rate of development of resistance to treatment, and/or a reduction in tumor invasiveness.
  • the compound of Formula (I), or a pharmaceutically acceptable salt thereof, or a pharmaceutically acceptable salt, or N-oxide thereof has the structure of Formula (II), or a pharmaceutically acceptable salt, or N-oxide thereof.
  • the compound of Formula (I), or a pharmaceutically acceptable salt thereof is a compound described in Table 1, or a pharmaceutically acceptable salt thereof.
  • Compounds disclosed herein are estrogen receptor modulators.
  • compounds disclosed herein have high specificity for the estrogen receptor and have desirable, tissue-selective pharmacological activities. Desirable, tissue-selective pharmacological activities include, but are not limited to, ER antagonist activity in breast cells and minimal or no ER agonist activity in uterine cells.
  • compounds disclosed herein are estrogen receptor degraders that display full estrogen receptor antagonist activity with negligible or minimal estrogen receptor agonist activity.
  • compounds disclosed herein are estrogen receptor degraders. In some embodiments, compounds disclosed herein are estrogen receptor antagonists. In some embodiments, compounds disclosed herein have minimal or negligible estrogen receptor agonist activity.
  • presented herein are compounds selected from active metabolites, tautomers, pharmaceutically acceptable solvates, pharmaceutically acceptable salts or prodrugs of a compound of Formula (I).
  • compositions comprising a therapeutically effective amount of a compound of Formula (I), or a pharmaceutically acceptable salt, or N-oxide thereof.
  • the pharmaceutical composition also contains at least one pharmaceutically acceptable inactive ingredient.
  • the pharmaceutical composition is formulated for intravenous injection, subcutaneous injection, oral administration, or topical administration.
  • the pharmaceutical composition is a tablet, a pill, a capsule, a liquid, a suspension, a gel, a dispersion, a suspension, a solution, an emulsion, an ointment, or a lotion.
  • the pharmaceutical composition further comprises one or more additional therapeutically active agents selected from: corticosteroids, anti-emetic agents, analgesics, anti-cancer agents, anti-inflammatories, kinase inhibitors, antibodies, HSP90 inhibitors, histone deacetylase (HDAC) inhibitors, poly ADP-ribose polymerase (PARP) inhibitors, and aromatase inhibitors.
  • additional therapeutically active agents selected from: corticosteroids, anti-emetic agents, analgesics, anti-cancer agents, anti-inflammatories, kinase inhibitors, antibodies, HSP90 inhibitors, histone deacetylase (HDAC) inhibitors, poly ADP-ribose polymerase (PARP) inhibitors, and aromatase inhibitors.
  • a method comprising administering a compound of Formula (I), or a pharmaceutically acceptable salt, or N-oxide thereof to a human with a diseases or condition that is estrogen sensitive, estrogen receptor meditated or estrogen receptor dependent.
  • the human is already being administered one or more additional therapeutically active agents other than a compound of Formula (I), or a pharmaceutically acceptable salt, or N-oxide thereof.
  • the method further comprises administering one or more additional therapeutically active agents other than a compound of Formula (I), or a pharmaceutically acceptable salt, or N-oxide thereof.
  • the one or more additional therapeutically active agents other than a compound of Formula (I), or a pharmaceutically acceptable salt, or N-oxide thereof are selected from: corticosteroids, anti-emetic agents, analgesics, anti-cancer agents, anti-inflammatories, kinase inhibitors, antibodies, HSP90 inhibitors, histone deacetylase (HDAC) inhibitors, and aromatase inhibitors.
  • the methods further comprise administering to the mammal radiation therapy.
  • the compound of the methods is administered prior to or following surgery.
  • the methods comprise administering to the mammal at least one additional anti-cancer agent.
  • the cancer is breast cancer, ovarian cancer, endometrial cancer, prostate cancer, uterine cancer, cervical cancer or lung cancer.
  • the cancer is breast cancer.
  • the cancer is a hormone dependent cancer.
  • the cancer is an estrogen receptor dependent cancer.
  • the cancer is an estrogen-sensitive cancer.
  • the cancer is resistant to anti-hormonal treatment.
  • the cancer is an estrogen-sensitive cancer or an estrogen receptor dependent cancer that is resistant to anti-hormonal treatment.
  • the anti-hormonal treatment includes treatment with at least one agent selected from tamoxifen, fulvestrant, steroidal aromatase inhibitors, and non-steroidal aromatase inhibitors.
  • the methods further comprise administering to the mammal radiation therapy.
  • the compound of the methods is administered prior to or following surgery.
  • the methods comprise administering to the mammal at least one additional anti-cancer agent.
  • methods of treating hormone receptor positive metastatic breast cancer in a postmenopausal woman with disease progression following anti-estrogen therapy comprising administering to the woman an estrogen receptor degrading compound of Formula (I), or a pharmaceutically acceptable salt, or N-oxide thereof.
  • the methods further comprise administering to the mammal radiation therapy.
  • the compound of the methods is administered prior to or following surgery.
  • the methods comprise administering to the mammal at least one additional anti-cancer agent.
  • methods of treating a hormonal dependent benign or malignant disease of the breast or reproductive tract in a mammal comprising administering to the mammal an effective amount of a compound of Formula (I), a pharmaceutically acceptable salt, or N-oxide thereof.
  • the benign or malignant disease is breast cancer.
  • the methods further comprise administering to the mammal radiation therapy.
  • the compound of the methods is administered prior to or following surgery.
  • the methods comprise administering to the mammal at least one additional anti-cancer agent.
  • a compound of Formula (I), or a pharmaceutically acceptable salt, or N-oxide thereof is used to treat hormone receptor positive metastatic breast cancer in a postmenopausal woman with disease progression following anti-estrogen therapy.
  • the compound is used to treat a hormonal dependent benign or malignant disease of the breast or reproductive tract in a mammal.
  • a method of reducing ER activation in a mammal comprising administering to the mammal at least one compound having the structure of Formula (I), or a pharmaceutically acceptable salt, or N-oxide thereof.
  • the method comprises reducing ER activation in breast cells, ovarian cells, colon cells, prostate cells, endometrial cells, or uterine cells in the mammal.
  • the method of reducing ER activation in the mammal comprises reducing the binding of estrogens to estrogen receptors in the mammal
  • the method of reducing ER activation in the mammal comprises reducing ER concentrations in the mammal.
  • the disease or condition is breast cancer, ovarian cancer, colon cancer, prostate cancer, endometrial cancer, or uterine cancer.
  • the disease or condition is bone cancer, breast cancer, colorectal cancer, endometrial cancer, prostate cancer, ovarian cancer, uterine cancer, cervical cancer, lung cancer, alcoholism, migraine, aortic aneurysm, susceptibility to myocardial infarction, aortic valve sclerosis, cardiovascular disease, coronary artery disease, hypertension, deep vein thrombosis, Graves' Disease, arthritis, multiple sclerosis, cirrhosis, hepatitis B, chronic liver disease, bone density, cholestasis, hypospadias, obesity, osteoarthritis, osteopenia, osteoporosis, Alzheimer's disease, Parkinson's disease, migraine, vertigo, anorexia nervosa, attention deficit hyperactivity disorder (ADHD), dementia, major depressive disorder, psychosis, age of menarche, endometriosis, or infertility in a mammal.
  • ADHD attention deficit hyperactivity disorder
  • dementia major depressive disorder
  • psychosis
  • a compound of Formula (I), or a pharmaceutically acceptable salt, or N-oxide thereof in the treatment or prevention of diseases or conditions that are estrogen sensitive, estrogen receptor dependent or estrogen receptor mediated.
  • the disease or condition is described herein.
  • the mammal is a human.
  • compounds provided herein are used to diminish, reduce, or eliminate the activity of estrogen receptors.
  • Estrogen receptor alpha (ER- ⁇ ; NR3A1) and estrogen receptor beta (ER- ⁇ ; NR3A2) are steroid hormone receptors, which are members of the large nuclear receptor superfamily. Nuclear receptors share a common modular structure, which minimally includes a DNA binding domain (DBD) and a ligand binding domain (LBD). Steroid hormone receptors are soluble, intracellular proteins that act as ligand-regulated transcription factors. Vertebrates contain five closely related steroid hormone receptors (estrogen receptor, androgen receptor, progesterone receptor, glucocorticoid receptor, mineralcorticoid receptor), which regulate a wide spectrum of reproductive, metabolic and developmental activities. The activities of ER are controlled by the binding of endogenous estrogens, including 17 ⁇ -estradiol and estrones.
  • the ER- ⁇ gene is located on 6q25.1 and encodes a 595 AA protein.
  • the ER- ⁇ gene resides on chromosome 14q23.3 and produces a 530 AA protein.
  • each of these genes can give rise to multiple isoforms.
  • these receptors contain an N-terminal (A/B) domain, a hinge (D) domain that links the C and E domains, and a C-terminal extension (F domain) (Gronemeyer and Laudet; Protein Profile 2: 1173-1308, 1995).
  • the ligand binding pocket of steroid hormone receptors is deeply buried within the ligand binding domain. Upon binding, the ligand becomes part of the hydrophobic core of this domain. Consequently most steroid hormone receptors are instable in the absence of hormone and require assistance from chaperones, such as Hsp90, in order to maintain hormone-binding competency. The interaction with Hsp90 also controls nuclear translocation of these receptors.
  • Ligand-binding stabilizes the receptor and initiates sequential conformational changes that release the chaperones, alter the interactions between the various receptor domains and remodel protein interaction surfaces that allow these receptors to translocate into the nucleus, bind DNA and engage in interactions with chromatin remodeling complexes and the transcriptional machinery.
  • ER can interact with Hsp90, this interaction is not required for hormone binding and, dependent on the cellular context, apo-ER can be both cytoplasmic and nuclear. Biophysical studies indicated that DNA binding rather than ligand binding contributes to the stability of the receptor (Greenfield et al., Biochemistry 40: 6646-6652, 2001).
  • ER can interact with DNA either directly by binding to a specific DNA sequence motif called estrogen response element (ERE) (classical pathway), or indirectly via protein-protein interactions (nonclassical pathway) (Welboren et al., Endocrine-Related Cancer 16: 1073-1089, 2009).
  • EAE estrogen response element
  • nonclassical pathway protein-protein interactions
  • Both types of ER DNA interactions can result in gene activation or repression dependent on the transcriptional coregulators that are recruited by the respective ER-ERE complex (Klinge, Steroid 65: 227-251, 2000).
  • the recruitment of coregulators is primarily mediated by two protein interaction surfaces, the AF2 and AF1.
  • AF2 is located in the ER E-domain and its conformation is directly regulated by the ligand (Brzozowski et al., Nature 389: 753-758, 1997).
  • Full agonists appear to promote the recruitment of co-activators, whereas weak agonists and antagonists facilitate the binding of co-repressors.
  • the regulation of protein with the AF 1 is less well understood but can be controlled by serine phosphorylation (Ward and Weigel, Biofactors 35: 528-536, 2009).
  • One of the involved phosphorylation sites appears to control the transcriptional activity of ER in the presence of antagonists such as tamoxifen, which plays an important role in the treatment of breast cancer. While full agonists appear to arrest ER in certain conformation, weak agonists tend to maintain ER in equilibrium between different conformations, allowing cell-dependent differences in co-regulator repertoires to modulate the activity of ER in a cell-dependent manner (Tamrazi et al., Mol. Endocrinol. 17: 2593-2602, 2003).
  • Interactions of ER with DNA are dynamic and include, but are not limited to, the degradation of ER by the proteasome (Reid et al., Mol Cell 11: 695-707, 2003).
  • the degradation of ER with ligands provides an attractive treatment strategy for disease or conditions that estrogen-sensitive and/or resistant to available anti-hormonal treatments.
  • ER signaling is crucial for the development and maintenance of female reproductive organs including breasts, ovulation and thickening of the endometrium. ER signaling also has a role in bone mass, lipid metabolism, cancers, etc. About 70% of breast cancers express ER- ⁇ (ER- ⁇ positive) and are dependent on estrogens for growth and survival. Other cancers also are thought to be dependent on ER- ⁇ signaling for growth and survival, such as for example ovarian and endometrial cancers.
  • the ER- ⁇ antagonist tamoxifen has been used to treat early and advanced ER- ⁇ positive breast cancer in both pre- and post-menopausal women.
  • Fulvestrant a steroid-based ER antagonist is used to treat breast cancer in women which has have progressed despite therapy with tamoxifen.
  • Steroidal and non-steroidal aromatase inhibitors are also used to treat cancers in humans.
  • the steroidal and non-steroidal aromatase inhibitors block the production of estrogen from androstenedione and testosterone in post-menopausal women, thereby blocking ER dependent growth in the cancers.
  • progressive ER positive breast cancer is treated in some cases with a variety of other chemotherapeutics, such as for example, the anthracylines, platins, taxanes.
  • ER positive breast cancers that harbor genetic amplication of the ERB-B/HER2 tyrosine kinase receptor are treated with the monoclonal antibody trastuzumab (HerceptinTM) or the small molecule pan-ERB-B inhibitor lapatinib.
  • HerceptinTM monoclonal antibody trastuzumab
  • lapatinib small molecule pan-ERB-B inhibitor lapatinib.
  • SERMs selective estrogen receptor modulators
  • SERMs described herein are selective estrogen receptor degraders (SERDs).
  • SELDs selective estrogen receptor degraders
  • in cell-based assays the compounds described herein result in a reduction in steady state ER- ⁇ levels (i.e. ER degradation) and are useful in the treatment of estrogen sensitive diseases or conditions and/or diseases or conditions that have developed resistant to anti-hormonal therapies.
  • compounds disclosed herein minimize levels of the estrogen receptor in the nucleus.
  • compounds disclosed herein are useful in the treatment of breast cancer, either alone or in combination with other agent agents that modulate other critical pathways in breast cancer, including but not limited to those that target IGF1R, EGFR, erB-B2 and 3 the PI3K/AKT/mTOR axis, HSP90, PARP or histone deacetylases.
  • compounds disclosed herein are useful in the treatment of breast cancer, either alone or in combination with other agent used to treat breast cancer, including but not limited to aromatase inhibitors, anthracylines, platins, nitrogen mustard alkylating agents, taxanes.
  • Illustrative agent used to treat breast cancer include, but are not limited to, paclitaxel, anastrozole, exemestane, cyclophosphamide, epirubicin, fulvestrant, letrozole, gemcitabine, trastuzumab, pegfilgrastim, filgrastim, tamoxifen, docetaxel, toremifene, vinorelbine, capecitabine, ixabepilone, as well as others described herein.
  • ER-related diseases or conditions include ER- ⁇ dysfunction is also associated with cancer (bone cancer, breast cancer, colorectal cancer, endometrial cancer, prostate cancer, ovarian and uterine cancer), leiomyoma (uterine leiomyoma), central nervous system (CNS) defects (alcoholism, migraine), cardiovascular system defects (aortic aneurysm, susceptibility to myocardial infarction, aortic valve sclerosis, cardiovascular disease, coronary artery disease, hypertension), hematological system defects (deep vein thrombosis), immune and inflammation diseases (Graves' Disease, arthritis, multiple sclerosis, cirrhosis), susceptibility to infection (hepatitis B, chronic liver disease), metabolic defects (bone density, cholestasis, hypospadias, obesity, osteoarthritis, osteopenia, osteoporosis), neurological defects (Alzheimer's disease, Parkinson's disease, migraine, vertigo),
  • compounds disclosed herein are used to treat cancer in a mammal.
  • the cancer is breast cancer, ovarian cancer, endometrial cancer, prostate cancer, uterine cancer, cervical cancer or lung cancer.
  • the cancer is breast cancer.
  • the cancer is a hormone dependent cancer.
  • the cancer is an estrogen receptor dependent cancer.
  • the cancer is an estrogen-sensitive cancer.
  • the cancer is resistant to anti-hormonal treatment.
  • the cancer is an estrogen-sensitive cancer or an estrogen receptor dependent cancer that is resistant to anti-hormonal treatment.
  • anti-hormonal treatment includes treatment with at least one agent selected from tamoxifen, fulvestrant, steroidal aromatase inhibitors, and non-steroidal aromatase inhibitors-resistant.
  • compounds disclosed herein are used to treat hormone receptor positive metastatic breast cancer in a postmenopausal woman with disease progression following anti-estrogen therapy.
  • compounds disclosed herein are used to treat a hormonal dependent benign or malignant disease of the breast or reproductive tract in a mammal.
  • the benign or malignant disease is breast cancer.
  • compounds disclosed herein are used to treat cancer in a mammal, wherein the mammal is chemotherapy-na ⁇ ve.
  • compounds disclosed herein are used to treat cancer in a mammal, wherein the mammal is being treated for cancer with at least one anti-cancer agent.
  • the cancer is a hormone refractory cancer.
  • compounds disclosed herein are used in the treatment of endometriosis in a mammal.
  • compounds disclosed herein are used in the treatment of leiomyoma in a mammal.
  • the leiomyoma is an uterine leiomyoma, esophageal leiomyoma, cutaneous leiomyoma or small bowel leiomyoma.
  • compounds disclosed herein are used in the treatment of fibroids in a mammal (e.g. uterine fibroids).
  • Compounds of Formula (I), including pharmaceutically acceptable salts, prodrugs, active metabolites and pharmaceutically acceptable solvates thereof, are estrogen receptor modulators.
  • the compounds described herein are estrogen receptor degraders.
  • the compounds described herein are estrogen receptor antagonists.
  • the compounds described herein are estrogen receptor degraders and estrogen receptor antagonists with minimal or no estrogen receptor agonist activity.
  • compounds disclosed herein are estrogen receptor degraders and estrogen receptor antagonists that exhibit: minimal or no estrogen receptor agonism; and/or anti-proliferative activity against breast cancer, ovarian cancer, endometrial cancer, cervical cancer cell lines; and/or maximal anti-proliferative efficacy against breast cancer, ovarian cancer, endometrial cancer, cervical cell lines in-vitro; and/or minimal agonism in the human endometrial (Ishikawa) cell line; and/or no agonism in the human endometrial (Ishikawa) cell line; and/or minimal or no agonism in the immature rat uterine assay in-vivo; and/or inverse agonism in the immature rat uterine assay in-vivo; and/or anti-tumor activity in breast cancer, ovarian cancer, endometrial cancer, cervical cancer cell lines in xenograft assays in-vivo or other rodent models of these cancers.
  • substituents are selected from among from a subset of the listed alternatives.
  • Z is —OH.
  • Z is —OR 10 .
  • Z is —OH, —OCH 3 , or —OCH 2 CH 3 .
  • R 6 is H, —CH 3 , F, or Cl. In some embodiments, R 6 is H.
  • R 7 is H, —CH 3 , F, or Cl. In some embodiments, R 7 is H.
  • R 9 is H, C 1 -C 6 alkyl, or C 1 -C 6 -fluoroalkyl. In some embodiments, R 9 is H or C 1 -C 6 alkyl. In some embodiments, R 9 is H.
  • R 3 may be present on any open position of the indazole ring system.
  • each R 3 is independently halogen, C 1 -C 4 alkyl, or C 1 -C 4 -fluoroalkyl.
  • each R 3 is independently F, Cl, or —CH 3 .
  • each R 4 is independently halogen, —CN, —OH, —OR 9 , —S( ⁇ O) 2 R 10 , C 1 -C 4 alkyl, C 1 -C 4 -fluoroalkyl, C 1 -C 4 heteroalkyl, C 1 -C 4 -fluoroalkoxy, or C 1 -C 4 alkoxy.
  • each R 4 is independently halogen, —CN, —OH, —S( ⁇ O) 2 CH 3 , —S( ⁇ O) 2 CH 2 CH 3 , —CH 3 , —CH 2 CH 3 , —CF 3 , —CH 2 OH, —OCF 3 , —OCH 3 , or —OCH 2 CH 3 .
  • each R 4 is independently F, Cl, —CN, —OH, —CH 3 , —CH 2 CH 3 , —CF 3 , —CH 2 OH, —OCH 3 , or —OCH 2 CH 3 .
  • each R 4 is independently F or Cl.
  • each R 5 is independently halogen, C 1 -C 4 alkyl, or C 1 -C 4 -fluoroalkyl. In some embodiments, each R 5 is independently F, Cl, or —CH 3 .
  • m is 0 or 1. In some embodiments, m is 0. In some embodiments, m is 1.
  • n is 0, 1, or 2. In some embodiments, n is 0. In some embodiments, n is 1. In some embodiments, n is 2.
  • p is 0 or 1. In some embodiments, p is 0. In some embodiments, p is 1.
  • R 2 is C 1 -C 4 alkyl, C 1 -C 4 -fluoroalkyl, C 1 -C 4 deuteroalkyl, C 3 -C 6 cycloalkyl, or —C 1 -C 4 alkylene-W; W is hydroxy, halogen, CN, C 1 -C 4 alkoxy, or C 3 -C 6 cycloalkyl.
  • R 2 is C 1 -C 4 alkyl, C 1 -C 4 -fluoroalkyl, or C 1 -C 4 deuteroalkyl.
  • R 2 is C 1 -C 4 alkyl.
  • R 2 is —CH 3 , —CH 2 CH 3 , —CH 2 CH 2 CH 3 , —CH(CH 3 ) 2 , —CH 2 CH 2 CH 2 CH 3 , —CH 2 CH(CH 3 ) 2 , —CH 2 F, —CHF 2 , —CF 3 , —CH 2 CF 3 , —CD 3 , —CH 2 CD 3 , —CD 2 CD 3 , cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, —CH 2 —W, or —CH 2 CH 2 —W; W is hydroxy, F, Cl, —CN, —OCH 3 , —OCH 2 CH 3 , —OCH 2 CH 2 CH 3 , —OCH(CH 3 ) 2 , cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl.
  • W is hydroxy, F, Cl, —CN, cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl.
  • R 2 is —CH 3 , —CH 2 CH 3 , —CH 2 CH 2 CH 3 , —CH(CH 3 ) 2 , —CH 2 CH 2 CH 2 CH 3 , —CH 2 CH(CH 3 ) 2 , —CH 2 F, —CHF 2 , —CF 3 , —CH 2 CF 3 , —CD 3 , —CH 2 CD 3 , —CD 2 CD 3 , —CH 2 —W, or —CH 2 CH 2 —W.
  • R 2 is —CH 3 , —CH 2 CH 3 , —CH 2 F, —CHF 2 , —CF 3 , —CH 2 CF 3 , —CD 3 , —CD 2 CD 3 , —CH 2 CD 3 , cyclopropyl or cyclobutyl.
  • R 2 is —CH 2 CH 3 , —CH 2 CF 3 , —CD 2 CD 3 , or —CH 2 CD 3 .
  • R 2 is —CH 2 CH 3 , —CD 2 CD 3 , or —CH 2 CD 3 .
  • R 2 is —CH 2 CH 3 or cyclobutyl.
  • R 2 is —CH 2 CH 3 .
  • each R 3 is independently F, Cl, or —CH 3 ; and m is 0 or 1.
  • the compound of Formula (I) has the structure of Formula (II), or a pharmaceutically acceptable salt, or N-oxide thereof:
  • R 2 is —CH 3 , —CH 2 CH 3 , —CH 2 CH 2 CH 3 , —CH(CH 3 ) 2 , —CH 2 CH 2 CH 2 CH 3 , —CH 2 CH(CH 3 ) 2 , —CH 2 F, —CHF 2 , —CF 3 , —CH 2 CF 3 , —CD 3 , —CD 2 CD 3 , cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, —CH 2 —W, or —CH 2 CH 2 —W; W is hydroxy, F, Cl, —CN, —OCH 3 , —OCH 2 CH 3 , —OCH 2 CH 2 CH 3 , —OCH(CH 3 ) 2 , cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl.
  • W is hydroxy, F, Cl, —CN, cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl; each R 4 is independently F, Cl, —CN, —OH, —CH 3 , —CF 3 , —OCF 3 , or —OCH 3 ; m is 0 or 1; n is 0, 1, or 2; and p is 0.
  • R 2 is —CH 2 CH 3 or cyclobutyl; each R 4 is independently F, Cl, —CN, —OH, —CH 3 , —CH 2 CH 3 , —CF 3 , —CH 2 OH, —OCF 3 , —OCH 3 , or —OCH 2 CH 3 ; m is 0 or 1; n is 0, 1, or 2; and p is 0.
  • R 2 is —CH 2 CH 3 ; each R 4 is independently F, Cl, —CN, —OH, —CH 3 , —CH 2 CH 3 , —CF 3 , —CH 2 OH, —OCF 3 , —OCH 3 , or —OCH 2 CH 3 ; m is 0; n is 0, 1, or 2; and p is 0.
  • the compound of Formula (I) or Formula (II) has the following structure, or a pharmaceutically acceptable salt thereof, or N-oxide thereof:
  • the compound of Formula (I) is a compound presented in Table 1, or a pharmaceutically acceptable salt, or N-oxide thereof:
  • the starting material used for the synthesis of the compounds of Formula (I) are either synthesized or obtained from commercial sources, such as, but not limited to, Sigma-Aldrich, Fluka, Acros Organics, Alfa Aesar, and the like.
  • the compounds described herein, and other related compounds having different substituents are synthesized using techniques and materials described herein or otherwise known, including those found in March, A DVANCED O RGANIC C HEMISTRY 4 th Ed., (Wiley 1992); Carey and Sundberg, A DVANCED O RGANIC C HEMISTRY 4 th Ed., Vols.
  • exemplary compounds of Formula (I) are prepared as outlined in the following Schemes.
  • the protecting group is tetrahydro-2H-pyran (THP).
  • the conditions for nitrogen protection require 3,4-dihydro-2H-pyran (DHP), an organic acid and a suitable solvent.
  • the organic acid is pyridinium p-toluenesulfonate (PPTS) or p-toluenesulfonic acid (p-TSA or p-TsOH) and the suitable solvent is dichloromethane.
  • the reaction is performed at room temperature. Other conditions to protect the nitrogen of the starting material are known.
  • Sonogashira coupling reaction conditions include the use of a palladium catalyst and a copper halide salt.
  • the Sonogashira reaction conditions in the use of Pd(Ph 3 P) 2 Cl 2 , CuI, and triethylamine. In one embodiment, the reaction is performed at about 80° C. Other suitable reaction conditions are described in Rafael Chinchilla and Carmen Nájera (2007). Chem. Rev. 107 (3): 874-922.
  • the silyl protecting group of compounds of structure 3 is removed under suitable reaction conditions to provide compounds of structure 4.
  • the silyl protecting group is removed with potassium carbonate (K 2 CO 3 ) in methanol.
  • the silyl protecting group is removed with tetrabutylammoniumfluoride (TBAF) in tetrahydrofuran.
  • K 2 CO 3 potassium carbonate
  • TBAF tetrabutylammoniumfluoride
  • acetylenes of Structure 4 are reacted with R 2 —X under basic condition to prepare compounds of Structure 5.
  • R 2 is C 1 -C 6 alkyl or C 1 -C 6 -fluoroalkyl or C 1 -C 4 alkoxy or C 1 -C 4 -fluoroalkoxy or C 3 -C 6 cycloalkyl, or the like
  • X is a suitable leaving group.
  • R 2 moieties are installed by other suitable conditions.
  • compounds of structure 2 are coupled with an alkynyl-trimethylsilane or a terminal-alkyne under Sonogashira reaction conditions to provide compounds of structure 5.
  • the coupling of an alkynyl-trimethylsilane with compounds of structure 2 includes the use of a base (e.g. cesium carbonate), a palladium catalyst (e.g. Pd(OAc) 2 , dppf) and a copper halide salt (e.g. CuI) in a suitable solvent (e.g. dimethylacetamide), at elevated temperatures (e.g. about 80-90° C.).
  • the coupling of a terminal-alkyne with compounds of structure 2 includes the use of Pd(PPh 3 ) 2 Cl 2 ), CuI, and triethylamine, with the reaction performed with at elevated temperatures (e.g. about 80-120° C.).
  • the suitable reaction conditions include the use of organometallic reagent(s).
  • the suitable reaction conditions include the use of a palladium catalyst.
  • the suitable reaction conditions include the use of Pd(PhCN) 2 Cl 2 , K 2 CO 3 in dimethylformamide/water.
  • Other suitable reaction conditions include those described in Chengxiang Zhou and Richard C. Larock, Journal of Organic Chemistry, 2005, 70, 3765-3777; Chengxiang Zhou, Daniel E. Emrich, and Richard C. Larock Organic Letters 2003, 1579-1582; Tsutomu Konno, Ken-ichi Taku, Takashi Ishihara, Journal of Fluorine Chemistry 127 (2006) 966-972.
  • the protecting group of compounds of structure 8 is then removed under suitable reaction conditions to provide compounds of structure 9.
  • the suitable reaction conditions include the use of an acid.
  • the suitable reaction conditions include the use of hydrochloric acid, ethanol, with the reaction performed at about 70° C.
  • Hydrolysis of the ester group of compounds of structure 9 provides carboxylic acid compounds of structure 10.
  • the hydrolysis reaction includes the use of lithium hydroxide in a mixture of tetrahydrofuran and ethanol. Other hydrolysis reaction conditions are known.
  • compounds of structure 5 are reacted with phenyl halides of structure 6 and boronic acids of structure 11 under suitable reaction conditions to provide compounds of structure 12.
  • the suitable reaction conditions include the use of organometallic reagent(s).
  • the suitable organometallic reagent is a palladium catalyst.
  • the aldehyde of compounds of structure 12 is then transformed to an alkene under suitable reaction conditions to provide compounds of structure 8.
  • Suitable reaction conditions include a Horner-Wadsworth-Emmons olefination reaction or a Wittig olefination reaction conditions.
  • compounds of structure 5 are reacted with a borylating agent in the presence of a suitable catalyst to provide compounds of structure 13.
  • the suitable catalyst is an organometallic reagent such as a platinum catalyst.
  • the amount of catalyst impacts the rate of the reaction, but generally, not the yield or purity.
  • the solvent has a small impact on the rate of the reaction, but generally, not the yield or purity.
  • the temperature has a significant impact on the rate of the reaction, but generally, not the yield or purity.
  • a Suzuki cross-coupling is then performed with compounds of structure 13 and phenyl halides of structure 14 to provide compounds of structure 15.
  • 2 or 3 equivalents of base e.g.
  • Cs 2 CO 3 is used in the Suzuki cross-coupling.
  • 1.3 equivalents of base e.g. Cs 2 CO 3
  • the solvent has a significant impact on the rate and regioselectivity of this reaction.
  • dioxane, DME, or 2-MeTHF is used.
  • water content has a significant impact on the rate and regioselectivity of this the Suzuki cross-coupling.
  • a subsequent Suzuki cross-coupling is then performed between compounds of structure 15 and phenyl halides of structure 16 to provide compounds of structure 12.
  • each stereocenter exists independently in either the R or S configuration.
  • the compounds presented herein include all diastereomeric, enantiomeric, atropisomers, and epimeric forms as well as the appropriate mixtures thereof.
  • the compounds and methods provided herein include all cis, trans, syn, anti,
  • E
  • Z
  • isomers as well as the appropriate mixtures thereof.
  • Stereoisomers are obtained, if desired, by methods such as, stereoselective synthesis and/or the separation of stereoisomers by chiral chromatographic columns and/or use of optically active resolving agents. Jean Jacques, Andre Collet, Samuel H. Wilen, “Enantiomers, Racemates and Resolutions”, John Wiley And Sons, Inc., 1981.
  • the compounds presented herein are present as atropisomers.
  • Atropisomers refer to stereoisomers resulting from hindered rotation about single bonds where the steric strain barrier to rotation allows for the isolation of conformers.
  • Atropisomers display axial chirality. Separation of atropisomers is possible. In some embodiments, separation of atropisomers is possible by chiral resolution methods such as selective crystallization.
  • Atropisomers are optionally characterized by NMR or other suitable characterization means.
  • compositions described herein include the use of amorphous forms as well as crystalline forms (also known as polymorphs).
  • compounds described herein are in the form of pharmaceutically acceptable salts.
  • active metabolites of these compounds having the same type of activity are included in the scope of the present disclosure.
  • the compounds described herein can exist in unsolvated as well as solvated forms with pharmaceutically acceptable solvents such as water, ethanol, and the like.
  • the solvated forms of the compounds presented herein are also considered to be disclosed herein.
  • compounds described herein are prepared as prodrugs.
  • a “prodrug” refers to an agent that is converted into the parent drug in vivo. Prodrugs are often useful because, in some situations, they are easier to administer than the parent drug or they are bioavailable by oral administration or they have improved solubility in pharmaceutical compositions over the parent drug. In some embodiments, the design of a prodrug increases the effective water solubility.
  • An example, without limitation, of a prodrug is a compound described herein, which is administered as an ester (the “prodrug”) but then is metabolically hydrolyzed to provide the active entity.
  • a prodrug upon in vivo administration, a prodrug is chemically converted to the biologically, pharmaceutically or therapeutically active form of the compound.
  • sites on the aromatic ring portion of compounds of Formula (I) are susceptible to various metabolic reactions. Incorporation of appropriate substituents on the aromatic ring structures will reduce, minimize or eliminate this metabolic pathway. In specific embodiments, the appropriate substituent to decrease or eliminate the susceptibility of the aromatic ring to metabolic reactions is, by way of example only, a halogen, deuterium or an alkyl group.
  • the compounds described herein are labeled isotopically (e.g. with a radioisotope) or by another other means, including, but not limited to, the use of chromophores or fluorescent moieties, bioluminescent labels, or chemiluminescent labels.
  • the compounds described herein are metabolized upon administration to an organism in need to produce a metabolite that is then used to produce a desired effect, including a desired therapeutic effect.
  • “Pharmaceutically acceptable,” as used herein, refers to a material, such as a carrier or diluent, which does not abrogate the biological activity or properties of the compound, and is relatively nontoxic, i.e., the material is administered to an individual without causing undesirable biological effects or interacting in a deleterious manner with any of the components of the composition in which it is contained.
  • pharmaceutically acceptable salt refers to a formulation of a compound that does not cause significant irritation to an organism to which it is administered and does not abrogate the biological activity and properties of the compound.
  • pharmaceutically acceptable salts are obtained by reacting a compound of Formula (I) with an acid.
  • Pharmaceutically acceptable salts are also obtained by reacting a compound of Formula (I) with a base to form a salt.
  • compositions described herein are optionally formed as, and/or used as, pharmaceutically acceptable salts.
  • pharmaceutical acceptable salts include, but are not limited to: (1) acid addition salts, formed by reacting the free base form of the compound with a pharmaceutically acceptable: inorganic acid, such as, for example, hydrochloric acid, hydrobromic acid, sulfuric acid, phosphoric acid, metaphosphoric acid, and the like; or with an organic acid, such as, for example, acetic acid, propionic acid, hexanoic acid, cyclopentanepropionic acid, glycolic acid, pyruvic acid, lactic acid, malonic acid, succinic acid, malic acid, maleic acid, fumaric acid, trifluoroacetic acid, tartaric acid, citric acid, benzoic acid, 3-(4-hydroxybenzoyl)benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, 1,2-ethane
  • compounds described herein coordinate with an organic base, such as, but not limited to, ethanolamine, diethanolamine, triethanolamine, tromethamine, N-methylglucamine, dicyclohexylamine, tris(hydroxymethyl)methylamine.
  • compounds described herein form salts with amino acids such as, but not limited to, arginine, lysine, and the like.
  • Acceptable inorganic bases used to form salts with compounds that include an acidic proton include, but are not limited to, aluminum hydroxide, calcium hydroxide, potassium hydroxide, sodium carbonate, sodium hydroxide, and the like.
  • the compounds provided herein are prepared as lysine salts, sodium salts and other suitable amino acid salts. In some embodiments, the compounds provided herein are prepared as a sodium salt. In some embodiments, the compounds provided herein are prepared as an N-methylglucamine salt. In some embodiments, the compounds provided herein are prepared as a hydrochloride salt.
  • solvates contain either stoichiometric or non-stoichiometric amounts of a solvent, and are formed during the process of crystallization with pharmaceutically acceptable solvents such as water, ethanol, and the like. Hydrates are formed when the solvent is water, or alcoholates are formed when the solvent is alcohol. Solvates of compounds described herein are conveniently prepared or formed during the processes described herein. In addition, the compounds provided herein optionally exist in unsolvated as well as solvated forms.
  • N-oxides if appropriate
  • crystalline forms also known as polymorphs
  • pharmaceutically acceptable salts of compounds having the structure of Formula (I) as well as active metabolites of these compounds having the same type of activity.
  • alkyl refers to an aliphatic hydrocarbon group.
  • the alkyl group is saturated or unsaturated.
  • the alkyl moiety, whether saturated or unsaturated, is branched or straight chain.
  • the alkyl is selected from the group consisting of methyl, ethyl, propyl, iso-propyl, n-butyl, iso-butyl, sec-butyl, and t-butyl.
  • Typical alkyl groups include, but are in no way limited to, methyl, ethyl, propyl, isopropyl, butyl, isobutyl, sec-butyl, tertiary butyl, pentyl, neopentyl, hexyl, allyl, vinyl, acetylene, but-2-enyl, but-3-enyl, and the like.
  • Deuteroalkyl refers to an alkyl group where 1 or more hydrogen atoms of an alkyl are replaced with deuterium.
  • alkoxy refers to a (alkyl)O— group, where alkyl is as defined herein.
  • cycloalkyl refers to cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl or cyclohexenyl.
  • halo or, alternatively, “halogen” or “halide” means fluoro, chloro, bromo or iodo.
  • fluoroalkyl refers to an alkyl in which one or more hydrogen atoms are replaced by a fluorine atom.
  • heteroalkyl refers to an alkyl group in which one or more skeletal atoms of the alkyl are selected from an atom other than carbon, e.g., oxygen, nitrogen (e.g. —NH—, —N(alkyl)-, sulfur, or combinations thereof.
  • a heteroalkyl is a C 1 -C 6 heteroalkyl.
  • module means to interact with a target either directly or indirectly so as to alter the activity of the target, including, by way of example only, to enhance the activity of the target, to inhibit the activity of the target, to limit the activity of the target, or to extend the activity of the target.
  • modulator refers to a molecule that interacts with a target either directly or indirectly.
  • the interactions include, but are not limited to, the interactions of an agonist, partial agonist, an inverse agonist, antagonist, degrader, or combinations thereof.
  • a modulator is an antagonist.
  • a modulator is a degrader.
  • SERM selective estrogen receptor modulator
  • a SERM displays ER antagonist activity in some tissues and ER agonist activity in other tissues.
  • a SERM displays ER antagonist activity in some tissues and minimal or no ER agonist activity in other tissues.
  • a SERM displays ER antagonist activity in breast tissues, ovarian tissues, endometrial tissues, and/or cervical tissues but minimal or no ER agonist activity in uterine tissues.
  • antagonist refers to a small-molecule agent that binds to a nuclear hormone receptor and subsequently decreases the agonist induced transcriptional activity of the nuclear hormone receptor.
  • agonist refers to a small-molecule agent that binds to a nuclear hormone receptor and subsequently increases nuclear hormone receptor transcriptional activity in the absence of a known agonist.
  • inverse agonist refers to a small-molecule agent that binds to a nuclear hormone receptor and subsequently decreases the basal level of nuclear hormone receptor transcriptional activity that is present in the absence of a known agonist.
  • degrader refers to a small molecule agent that binds to a nuclear hormone receptor and subsequently lowers the steady state protein levels of said receptor.
  • a degrader as described herein lowers steady state estrogen receptor levels by at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90% or at least 95%.
  • SETD selective estrogen receptor degrader
  • estrogen dependent refers to diseases or conditions that would not occur, or would not occur to the same extent, in the absence of estrogen receptors.
  • ER-mediated refers to diseases or conditions that occur in the absence of estrogen receptors but can occur in the presence of estrogen receptors.
  • estrogen-sensitive refers to diseases or conditions that would not occur, or would not occur to the same extent, in the absence of estrogens.
  • cancer refers to an abnormal growth of cells which tend to proliferate in an uncontrolled way and, in some cases, to metastasize (spread).
  • types of cancer include, but is not limited to, solid tumors (such as those of the bladder, bowel, brain, breast, endometrium, heart, kidney, lung, uterus, lymphatic tissue (lymphoma), ovary, pancreas or other endocrine organ (thyroid), prostate, skin (melanoma or basal cell cancer) or hematological tumors (such as the leukemias and lymphomas) at any stage of the disease with or without metastases.
  • solid tumors such as those of the bladder, bowel, brain, breast, endometrium, heart, kidney, lung, uterus, lymphatic tissue (lymphoma), ovary, pancreas or other endocrine organ (thyroid), prostate, skin (melanoma or basal cell cancer) or hematological tumors (such as the leukemias and lymphomas) at any
  • cancers include, acute lymphoblastic leukemia, acute myeloid leukemia, adrenocortical carcinoma, anal cancer, appendix cancer, astrocytomas, atypical teratoid/rhabdoid tumor, basal cell carcinoma, bile duct cancer, bladder cancer, bone cancer (osteosarcoma and malignant fibrous histiocytoma), brain stem glioma, brain tumors, brain and spinal cord tumors, breast cancer, bronchial tumors, Burkitt lymphoma, cervical cancer, chronic lymphocytic leukemia, chronic myelogenous leukemia, colon cancer, colorectal cancer, craniopharyngioma, cutaneous T-Cell lymphoma, embryonal tumors, endometrial cancer, ependymoblastoma, ependymoma, esophageal cancer, ewing sarcoma family of tumors, eye cancer, reti
  • co-administration are meant to encompass administration of the selected therapeutic agents to a single patient, and are intended to include treatment regimens in which the agents are administered by the same or different route of administration or at the same or different time.
  • an “effective amount” or “therapeutically effective amount,” as used herein, refer to a sufficient amount of an agent or a compound being administered which will relieve to some extent one or more of the symptoms of the disease or condition being treated. The result includes reduction and/or alleviation of the signs, symptoms, or causes of a disease, or any other desired alteration of a biological system.
  • an “effective amount” for therapeutic uses is the amount of the composition comprising a compound as disclosed herein required to provide a clinically significant decrease in disease symptoms.
  • An appropriate “effective” amount in any individual case is optionally determined using techniques, such as a dose escalation study.
  • pharmaceutical combination means a product that results from the mixing or combining of more than one active ingredient and includes both fixed and non-fixed combinations of the active ingredients.
  • fixed combination means that the active ingredients, e.g. a compound of Formula (I), and a co-agent, are both administered to a patient simultaneously in the form of a single entity or dosage.
  • non-fixed combination means that the active ingredients, e.g. a compound of Formula (I), and a co-agent, are administered to a patient as separate entities either simultaneously, concurrently or sequentially with no specific intervening time limits, wherein such administration provides effective levels of the two compounds in the body of the patient.
  • cocktail therapy e.g. the administration of three or more active ingredients.
  • a “metabolite” of a compound disclosed herein is a derivative of that compound that is formed when the compound is metabolized.
  • active metabolite refers to a biologically active derivative of a compound that is formed when the compound is metabolized.
  • metabolized refers to the sum of the processes (including, but not limited to, hydrolysis reactions and reactions catalyzed by enzymes) by which a particular substance is changed by an organism. Thus, enzymes may produce specific structural alterations to a compound.
  • cytochrome P450 catalyzes a variety of oxidative and reductive reactions while uridine diphosphate glucuronyltransferases catalyze the transfer of an activated glucuronic-acid molecule to aromatic alcohols, aliphatic alcohols, carboxylic acids, amines and free sulphydryl groups.
  • Metabolites of the compounds disclosed herein are optionally identified either by administration of compounds to a host and analysis of tissue samples from the host, or by incubation of compounds with hepatic cells in vitro and analysis of the resulting compounds.
  • subject or “patient” encompasses mammals.
  • mammals include, but are not limited to humans, chimpanzees, apes, monkeys, cattle, horses, sheep, goats, swine, rabbits, dogs, cats, rats, mice, guinea pigs, and the like.
  • the mammal is a human.
  • treat include alleviating, abating or ameliorating at least one symptom of a disease or condition, preventing additional symptoms, inhibiting the disease or condition, e.g., arresting the development of the disease or condition, relieving the disease or condition, causing regression of the disease or condition, relieving a condition caused by the disease or condition, or stopping the symptoms of the disease or condition either prophylactically and/or therapeutically.
  • Suitable routes of administration include, but are not limited to, oral, parenteral (e.g., intravenous, subcutaneous, intramuscular), intranasal, buccal, topical, rectal, aerosol, ophthalmic, pulmonary, transmucosal, transdermal, vaginal, otic, nasal, and topical administration.
  • parenteral delivery includes intramuscular, subcutaneous, intravenous, intramedullary injections, as well as intrathecal, direct intraventricular, intraperitoneal, intralymphatic, and intranasal injections.
  • a compound as described herein is administered in a systemic manner. In certain other embodiments, a compound as described herein is administered in a local rather than systemic manner.
  • the compounds described herein are formulated into pharmaceutical compositions.
  • Pharmaceutical compositions are formulated in a conventional manner using one or more pharmaceutically acceptable inactive ingredients that facilitate processing of the active compounds into preparations that are used pharmaceutically. Proper formulation is dependent upon the route of administration chosen.
  • a summary of pharmaceutical compositions described herein is found, for example, in Remington: The Science and Practice of Pharmacy, Nineteenth Ed (Easton, Pa.: Mack Publishing Company, 1995); Hoover, John E., Remington's Pharmaceutical Sciences, Mack Publishing Co., Easton, Pa. 1975; Liberman, H. A. and Lachman, L., Eds., Pharmaceutical Dosage Forms, Marcel Decker, New York, N.Y., 1980; and Pharmaceutical Dosage Forms and Drug Delivery Systems, Seventh Ed. (Lippincott Williams & Wilkins 1999), herein incorporated by reference for such disclosure.
  • a pharmaceutical composition refers to a mixture of a compound of Formula (I), with other chemical components (i.e. pharmaceutically acceptable inactive ingredients), such as carriers, excipients, binders, filling agents, suspending agents, flavoring agents, sweetening agents, disintegrating agents, dispersing agents, surfactants, lubricants, colorants, diluents, solubilizers, moistening agents, plasticizers, stabilizers, penetration enhancers, wetting agents, anti-foaming agents, antioxidants, preservatives, or one or more combination thereof.
  • the pharmaceutical composition facilitates administration of the compound to a mammal.
  • compositions will include at least one compound of Formula (I), or a pharmaceutically acceptable salt thereof, as an active ingredient in free-acid or free-base form, or in a pharmaceutically acceptable salt form.
  • methods and pharmaceutical compositions described herein include the use of N-oxides (if appropriate), crystalline forms, amorphous phases, as well as active metabolites of these compounds having the same type of activity.
  • compounds described herein exist in unsolvated form or in solvated forms with pharmaceutically acceptable solvents such as water, ethanol, and the like.
  • the pharmaceutical formulations described herein include, but are not limited to, aqueous liquid dispersions, self-emulsifying dispersions, solid solutions, liposomal dispersions, aerosols, solid dosage forms, powders, immediate release formulations, controlled release formulations, fast melt formulations, tablets, capsules, pills, delayed release formulations, extended release formulations, enteric coated formulations, pulsatile release formulations, multiparticulate formulations, and mixed immediate and controlled release formulations.
  • the compound of Formula (I), or a pharmaceutically acceptable salt thereof is administered systemically.
  • the compound of Formula (I), or a pharmaceutically acceptable salt thereof is administered orally. All formulations for oral administration are in dosages suitable for such administration.
  • the solid dosage forms disclosed herein are in the form of a tablet, a pill, a powder, a capsule, solid dispersion, solid solution, bioerodible dosage form, controlled release formulations, pulsatile release dosage forms, multiparticulate dosage forms, beads, pellets, granules.
  • the pharmaceutical formulation is in the form of a powder.
  • the pharmaceutical formulation is in the form of a tablet.
  • the pharmaceutical formulation is in the form of a suspension tablet, a fast-melt tablet, a bite-disintegration tablet, a rapid-disintegration tablet, an effervescent tablet, or a caplet.
  • pharmaceutical formulation is in the form of a capsule.
  • the pharmaceutical solid oral dosage forms are formulated to provide a controlled release of the active compound.
  • Controlled release profiles include, for example, sustained release, prolonged release, pulsatile release, and delayed release profiles.
  • liquid formulation dosage forms for oral administration are in the form of aqueous suspensions selected from the group including, but not limited to, pharmaceutically acceptable aqueous oral dispersions, emulsions, solutions, elixirs, gels, and syrups. See, e.g., Singh et al., Encyclopedia of Pharmaceutical Technology, 2nd Ed., pp. 754-757 (2002).
  • compositions optionally take the form of tablets, lozenges, or gels formulated in a conventional manner.
  • a compound of Formula (I), or a pharmaceutically acceptable salt thereof is formulated into a pharmaceutical composition suitable for intramuscular, subcutaneous, or intravenous injection.
  • Parenteral injections involve either bolus injection and/or continuous infusion.
  • the compound of Formula (I), or a pharmaceutically acceptable salt thereof is administered intravenously. In some embodiments, the compound of Formula (I), or a pharmaceutically acceptable salt thereof, is administered subcutaneously.
  • the compound of Formula (I), or a pharmaceutically acceptable salt thereof is administered topically.
  • the compound of Formula (I), or a pharmaceutically acceptable salt thereof is formulated into a variety of topically administrable compositions, such as solutions, suspensions, lotions, gels, pastes, shampoos, scrubs, rubs, smears, medicated sticks, medicated bandages, balms, creams or ointments.
  • the compound of Formula (I), or a pharmaceutically acceptable salt thereof is administered topically to the skin of mammal.
  • the compound of Formula (I), is prepared as a transdermal dosage form.
  • a compound of Formula (I), or a pharmaceutically acceptable salt thereof in the manufacture of a medicament for treating a disease, disorder or conditions in which the activity of estrogen receptors contributes to the pathology and/or symptoms of the disease or condition.
  • the disease or condition is any of the diseases or conditions specified herein.
  • the compound of Formula (I), or a pharmaceutically acceptable salt thereof is used in the preparation of medicaments for the treatment of diseases or conditions in a mammal that would benefit from a reduction of estrogen receptor activity.
  • Methods for treating any of the diseases or conditions described herein in a mammal in need of such treatment involves administration of pharmaceutical compositions that include at least one compound of Formula (I), or a pharmaceutically acceptable salt, N-oxide, active metabolite, prodrug, or pharmaceutically acceptable solvate thereof, in therapeutically effective amounts to said mammal.
  • Therapeutically effective amounts depend on the severity and course of the disease or condition, previous therapy, the patient's health status, weight, and response to the drugs, and the judgment of the treating physician. Therapeutically effective amounts are optionally determined by methods including, but not limited to, a dose escalation clinical trial.
  • the effective amount of the compound of Formula (I) is: (a) systemically administered to the mammal; and/or (b) administered orally to the mammal; and/or (c) intravenously administered to the mammal; and/or (d) administered by injection to the mammal; and/or (e) administered topically to the mammal; and/or (f) administered non-systemically or locally to the mammal.
  • the methods of treatment comprise single administration of the effective amount of the compound, including further embodiments in which (i) the compound is administered once; (ii) the compound is administered to the mammal multiple times over the span of one day; (iii) continually; or (iv) continuously.
  • any of the aforementioned aspects are further embodiments comprising multiple administrations of the effective amount of the compound, including further embodiments in which (i) the compound is administered continuously or intermittently: as in a single dose; (ii) the time between multiple administrations is every 6 hours; (iii) the compound is administered to the mammal every 8 hours; (iv) the compound is administered to the mammal every 12 hours; (v) the compound is administered to the mammal every 24 hours.
  • the method comprises a drug holiday, wherein the administration of the compound is temporarily suspended or the dose of the compound being administered is temporarily reduced; at the end of the drug holiday, dosing of the compound is resumed.
  • the length of the drug holiday varies from 2 days to 1 year.
  • the compound is administered chronically, that is, for an extended period of time.
  • the dose of drug being administered is temporarily reduced or temporarily suspended for a certain length of time (i.e., a “drug holiday”).
  • doses employed for adult human treatment are typically in the range of 0.01 mg-5000 mg per day. In one aspect, doses employed for adult human treatment are from about 1 mg to about 1000 mg per day. In one embodiment, the desired dose is conveniently presented in a single dose or in divided doses administered simultaneously or at appropriate intervals, for example as two, three, four or more sub-doses per day. In one embodiment, the daily dosages appropriate for the compound of Formula (I), or a pharmaceutically acceptable salt thereof, described herein are from about 0.01 to about 50 mg/kg per body weight.
  • the pharmaceutical composition further comprises one or more anti-cancer agents.
  • a compound of Formula (I), or a pharmaceutically acceptable salt thereof is co-administered with a second therapeutic agent, wherein the compound of Formula (I), or a pharmaceutically acceptable salt thereof, and the second therapeutic agent modulate different aspects of the disease, disorder or condition being treated, thereby providing a greater overall benefit than administration of either therapeutic agent alone.
  • the overall benefit experienced by the patient is the additive effect of the two therapeutic agents or the patient may experience a synergistic benefit.
  • the multiple therapeutic agents are administered in any order or even simultaneously. If administration is simultaneous, the multiple therapeutic agents are, by way of example only, provided in a single, unified form, or in multiple forms (e.g., as a single pill or as two separate pills).
  • methods for treatment of estrogen receptor-dependent or estrogen receptor-mediated conditions or diseases comprises administration to a mammal a compound of Formula (I), or a pharmaceutically acceptable salt thereof, in combination with at least one additional therapeutic agent.
  • a compound of Formula (I), or a pharmaceutically acceptable salt thereof in combination with hormone blocking therapy, chemotherapy, radiation therapy, monoclonal antibodies, or combinations thereof.
  • the at least one additional therapeutic agent for use in combination with a compound of Formula (I), or a pharmaceutically acceptable salt thereof include one or more of the following: abiraterone; abarelix; adriamycin; actinomycin; acivicin; aclarubicin; acodazole hydrochloride; acronine; adozelesin; aldesleukin; alemtuzumab; allopurinol; alitretinoin; altretamine; ambomycin; ametantrone acetate; aminoglutethimide; aminolevulinic acid; amifostine; amsacrine; anastrozole; anthramycin; aprepitant; arsenic trioxide; asparaginase; asperlin; azacitidine; azetepa; azotomycin; batimastat; bendamustine hydrochloride; benzodepa; bevacizum
  • a compound of Formula (I), or a pharmaceutically acceptable salt thereof is used in combination with anti-emetic agents to treat nausea or emesis, which result from the use of a compound of Formula (I), anti-cancer agent(s) and/or radiation therapy.
  • a compound of Formula (I), or a pharmaceutically acceptable salt thereof is used in combination with an agent useful in the treatment of anemia or neutropenia.
  • a compound of Formula (I), or a pharmaceutically acceptable salt thereof is administered with corticosteroids.
  • a compound of Formula (I), or a pharmaceutically acceptable salt thereof is coadministered with analgesics.
  • a compound of Formula (I), or a pharmaceutically acceptable salt thereof is used in combination with radiation therapy (or radiotherapy).
  • Radiation therapy is the treatment of cancer and other diseases with ionizing radiation.
  • Radiation therapy is optionally used to treat localized solid tumors, such as cancers of the skin, tongue, larynx, brain, breast, prostate, colon, uterus and/or cervix. It is also optionally used to treat leukemia and lymphoma (cancers of the blood-forming cells and lymphatic system, respectively).
  • a technique for delivering radiation to cancer cells is to place radioactive implants directly in a tumor or body cavity.
  • This is called internal radiotherapy (brachytherapy, interstitial irradiation, and intracavitary irradiation are types of internal radiotherapy.)
  • internal radiotherapy brachytherapy, interstitial irradiation, and intracavitary irradiation are types of internal radiotherapy.
  • brachytherapy interstitial irradiation, and intracavitary irradiation are types of internal radiotherapy.
  • internal radiotherapy Using internal radiotherapy, the radiation dose is concentrated in a small area, and the patient stays in the hospital for a few days. Internal radiotherapy is frequently used for cancers of the tongue, uterus, prostate, colon, and cervix.
  • radiation or “ionizing radiation” include all forms of radiation, including but not limited to ⁇ , ⁇ , and ⁇ radiation and ultraviolet light.
  • Step 1 1-(Tetrahydro-2H-pyran-2-yl)-5-((trimethylsilyl)ethynyl)-1H-indazole
  • lithium bis(trimethylsilyl)amide has been employed as the base in THF at 0° C. followed by alkylation with alkyl-halide at reflux.
  • Step 1 4-Methyl-1-(tetrahydro-2H-pyran-2-yl)-5-((trimethylsilyl)ethynyl)-1H-indazole
  • Ethynyltrimethylsilane was added to the reaction, and the mixture was heated at 80° C. for 90 min and then cooled to room temperature.
  • the reaction mixture was filtered through Celite and the Celite was washed with ethyl acetate (100 mL).
  • the filtrate was washed (2 ⁇ 50 mL sat'd NaHCO 3 ), dried (Na 2 SO 4 ), and concentrated under reduced pressure.
  • the crude material was purified on a silica gel column to yield the desired compound.
  • Prop-2-yn-1-ol (8.4 g, 150 mmol) was added via syringe and the reaction mixture was stirred at 80° C. for 16 hours. Upon completion, the solvent was evaporated. The residue was diluted with dichloromethane (400 mL), washed with water (3 ⁇ 200 mL) and brine (200 mL), dried over anhydrous sodium sulfate, and concentrated in vacuo. The residue was further purified on silica gel column (1:100-1:20 EtOAc/petroleum ether) affording the title compound (11.1 g).
  • reaction mixture was diluted with ethyl acetate and filtered through Celite. The filtrate was concentrated in vacuo and the residue was purified by column chromatography on silica gel (0-10% ethyl acetate in petroleum ether) affording the title compound (4.3 g).
  • Step 2 5-(3,3-Difluoroprop-1-yn-1-yl)-1-(tetrahydro-2H-pyran-2-yl)-1H-indazole
  • Step 3 1-(Tetrahydro-2H-pyran-2-yl)-5-(3,3,3-trifluoroprop-1-yn-1-yl)-1H-indazole
  • Step 1 4-(4-Fluoro-1-(tetrahydro-2H-pyran-2-yl)-1H-indazol-5-yl)but-3-yn-1-ol
  • Step 2 5-(4-Chlorobut-1-yn-1-yl)-4-fluoro-1-(tetrahydro-2H-pyran-2-yl)-1H-indazole
  • reaction mixture was allowed to warm to room temperature and stirred overnight. Upon completion, the reaction mixture was washed with water (4 ⁇ 600 mL) and then brine (2 ⁇ 500 mL). The organic layer was dried over anhydrous sodium sulfate and filtered. Hexane and THF were distilled off at 75110° C. But-1-yn-1-yltrimethylsilane was distilled between 125 to 135° C. affording 91 g of a colorless liquid (61%).
  • reaction mixture was diluted with EtOAc (300 mL) and H 2 O (300 mL) and then filtered. The organic layer of the filtrate was separated, and the aqueous layer was extracted with EtOAc (3 ⁇ 150 mL). The combined organic layers were washed with brine (2 ⁇ 100 mL), dried over anhydrous Na 2 SO 4 , filtered and concentrated.
  • Nitrogen was bubbled into a solution of 5-bromo-4-fluoro-1-(tetrahydro-2H-pyran-2-yl)-1H-indazole (19.7 g, 65.9 mmol; Intermediate 18) and DMA (60 mL). After 5 min, CuI (1.25 g, 6.6 mmol), Pd(OAc) 2 (1.48 g, 6.6 mmol), dppf (3.66 g, 6.6 mmol), Cs 2 CO 3 (34.3 g, 105.4 mmol), and but-1-yn-1-yltrimethylsilane (11.6 g, 92.3 mmol; Intermediate 35) were added sequentially with continued N 2 bubbling. The resulting mixture was heated at 80° C.
  • Step 4 5-(But-1-yn-1-yl)-4-chloro-1-(tetrahydro-2,1-pyran-2-yl)-1H-indazole
  • But-1-yn-1-yltrimethylsilane (1.80 g, 14.3 mmol; Intermediate 35) was then added and the tube was sealed immediately.
  • the reaction mixture was heated in a microwave reactor at 120° C. for 3 hours.
  • Four of these reactions (4 ⁇ 2 g scale per run) were combined, mixed with water (100 mL), and extracted with ethyl acetate (2 ⁇ 100 mL).
  • the combined organic layers were dried over Na 2 SO 4 and concentrated in vacuo.
  • the residue was purified by column chromatography on silica gel (0-3% ethyl acetate in petroleum ether) affording the title compound (4.5 g, 90%), which was then triturated with petroleum ether (8 mL).
  • Step 5 5-(But-1-yn-1-yl)-7-fluoro-1-(tetrahydro-2H-pyran-2-yl)-1H-indazole
  • Nitrogen was bubbled into a solution of 5-bromo-7-fluoro-1-(tetrahydro-2H-pyran-2-yl)-1H-indazole (7.5 g, 25.33 mmol) and DMA (100 mL). After 5 min, CuI (241 mg, 1.27 mmol), Pd(OAc) 2 (284 mg, 1.27 mmol), dppf (704 mg, 1.27 mmol), K 2 CO 3 (4.89 g, 35.46 mmol), and but-1-yn-1-yltrimethylsilane (4.46 g, 35.46 mmol) were added sequentially with continued N 2 bubbling. The resulting mixture was heated at 80° C. for 10 h under N 2 .
  • the reaction mixture was diluted with EtOAc (250 mL) and H 2 O (200 mL) and filtered. The organic layer of the filtrate was separated, and the aqueous layer was extracted with EtOAc (2 ⁇ 100 mL). The combined organic layers were washed with brine (3 ⁇ 50 mL), dried over anhydrous Na 2 SO 4 , concentrated, and purified by silica gel chromatography (1:30 EtOAc/petroleum ether) to give the pure product as a yellow solid (3.6 g) and an impure product (2 g; further purified to give additional 1.47 g).
  • the reaction mixture was diluted with EtOAc (350 mL) and H 2 O (300 mL) and filtered. The organic layer of the filtrate was separated, and the aqueous layer was extracted with EtOAc (2 ⁇ 50 mL). The combined organic layers were washed with brine (3 ⁇ 100 mL), dried over anhydrous Na 2 SO 4 , and concentrated in vacuo. The residue was purified on a silica gel column using EtOAc in petroleum ether (1:30) to afford the title compound as a yellow solid (3.9 g, yield 69.1%).
  • Step 2 5-(3-Cyclopropylprop-1-yn-1-yl)-1-(tetrahydro-2,1-pyran-2-yl)-1H-indazole
  • the reaction mixture was cooled to room temperature, diluted with EtOAc (100 mL) and H 2 O (100 mL) and filtered. The organic layer of the filtrate was separated, and the aqueous layer was extracted with additional EtOAc (2 ⁇ 50 mL). The combined organic layers were washed with brine (3 ⁇ 100 mL), dried over anhydrous Na 2 SO 4 , and concentrated in vacuo. The residue was purified on a silica gel column using 0-10% EtOAc in petroleum ether to afford the title compound as yellow oil (4.8 g, 71%).
  • 3,4-Dihydro-2H-pyran (1.1-10 equiv) was added to a solution of the appropriate halo-indazole (1.0 equiv), PPTS (or pTsOH, 0.05-0.3 equiv), and dichloromethane ( ⁇ 2 mL/mmol) at room temperature.
  • the reaction was stirred under N 2 for 6-48 h (until complete by TLC or LCMS), quenched with water, and then extracted with dichloromethane. The extracts were dried, filtered, concentrated, and purified by silica gel chromatography to give the protected halo-indazole.
  • the reaction was allowed to cool to room temperature, diluted with ethyl acetate and water, and then filtered through Celite. The aqueous layer was separated and extracted with ethyl acetate. The organics were combined, dried, filtered, concentrated, and then purified by silica gel column chromatography to give the alkynyl-indazole.
  • Alternate bases include K 2 CO 3 and CsF; Alternate ligands include 1,3-Bis(2,4,6-trimethylphenyl)imidazolium chloride and Ph 3 P; Alternate catalysts include Pd(PPh 3 ) 4 and PdCl 2 (PPh 3 ) 2 ; Alternate solvents include THF and pyrrolidine. Water content seems to impact the rate of this reaction: when anhydrous Cs 2 CO 3 and anhydrous solvent were used, 1% water (v/v with respect to solvent) was added to the reaction, and when the Cs 2 CO 3 and/or solvent were not anhydrous, no water was added.
  • Alternate catalysts include PdCl 2 (PPh 3 ) 2 ; Cosolvents include triethylamine and pyrrolidine; When employing bromo-heterocycles, the reaction temperature was increased (80-120° C.).
  • reaction was allowed to cool to room temperature and then either 1) taken directly into Step 2; 2) concentrated to give a crude residue [usually a foam]; or 3) concentrated and purified by silica gel chromatography to give the pure bis(pinacolato)diboryl-alkene.
  • the reaction was then either 1) taken directly into Step 3; or 2) processed to isolate the 1-aryl-2-(pinacolato)boryl-alkene: [The reaction was diluted with ethyl ether (or ethyl acetate) and washed with water (1-3 times). The aqueous phases were back extracted with ethyl ether (or ethyl acetate). The extracts were combined, dried, filtered, concentrated and then purified by silica gel chromatography].
  • Step 3 Cross-coupling of the 1-aryl-2-(pinacolato)boryl-alkene
  • the reaction was allowed to cool to room temperature, diluted with ethyl ether (or ethyl acetate), and washed with water (1-3 times). The aqueous phases were back extracted with ethyl ether (or ethyl acetate). The extracts were combined, dried, filtered, concentrated and then purified by silica gel chromatography to give the desired tetra-substituted alkene.
  • DBU 1,8-Diazabicyclo[5.4.0]undec-7-ene
  • Note 1 Most commonly, a 2M solution of aqueous LiOH was used, or the LiOH was dissolved in a minimum amount of water. In some instances, NaOH or KOH was used. Note 2: In some instances, a single solvent (ethanol, dioxane, or tetrahydrofuran) was used. Note 3: Alternate work-up procedures have been employed including: i) the use of sat'd NH 4 Cl in place of aqueous HCl and ii) removal of the organic solvent by rotary evaporation prior to acid quench.
  • Step 1 (E)-Ethyl 3-(4-((E)-2-phenyl-1-(1-(tetrahydro-2H-pyran-2-yl)-1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylate
  • Step 2 (E)-Ethyl 3-(4-((E)-1-(1H-indazol-5-yl)-2-phenylbut-1-en-1-yl)phenyl)acrylate
  • Compound 95 was prepared following the procedure outlined for Compound 94.
  • Step 1 (E)-Ethyl 3-(4-((E)-2-(3-hydroxyphenyl)-1-(1-(tetrahydro-2H-pyran-2-yl)-1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylate
  • Step 2 (E)-Ethyl 3-(4-((E)-2-(3-butoxyphenyl)-1-(1-(tetrahydro-2H-pyran-2-yl)-1H-indazol-5-yl)but-1-en-1-yl)phenypacrylate
  • Step 3 (E)-3-(4-((E)-2-(3-Butoxyphenyl)-1-(1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylic acid
  • Step 1 (E)-Ethyl 3-(4-((E)-2-(2-(methylthio)phenyl)-1-(1-(tetrahydro-2H-pyran-2-yl)-1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylate
  • Step 2 (E)-Ethyl 3-(4-((E)-2-(2-(methylsulfonyl)phenyl)-1-(1-(tetrahydro-2,1-pyran-2-yl)-1H-indazol-5-yl)but-1-en-1-yl)phenypacrylate
  • Step 3 (E)-3-(4-((E)-1-(1H-Indazol-5-yl)-2-(2-(methylsulfonyl)phenyl)but-1-en-1-yl)phenyl)acrylic acid
  • Step 1 (E)-4-(2-Phenyl-1-(1-(tetrahydro-2H-pyran-2-yl)-1H-indazol-5-yl)but-1-en-1-yl)benzaldehyde
  • Step 2 (E)-Ethyl 2-methyl-3-(4-((E)-2-phenyl-1-(1-(tetrahydro-2H-pyran-2-yl)-1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylate
  • Step 3 (E)-3-(4-((E)-1-(1H-Indazol-5-yl)-2-phenylbut-1-en-1-yl)phenyl)-2-methylacrylic acid
  • Step 1 (E)-4-(2-(2-Chloro-4-fluorophenyl)-1-(1-(tetrahydro-2H-pyran-2-yl)-1,1-indazol-5-yl)but-1-en-1-yl)benzaldehyde
  • Aqueous KOH solution (4M, 275 mL, 1100 mmol) and 2-chloro-4-fluoroiodobenzene (70.6 g, 275 mmol) were added.
  • the reaction was degassed with 3 vacuum/N 2 cycles, heated at 75° C. (internal temperature; oil bath at 90° C.) for 7 h under N 2 , and then allowed to cool to room temperature. The layers were separated, and the organic layer was washed with brine (800 mL), dried over sodium sulfate, filtered, and concentrated.
  • Step 2 (E)-Ethyl 3-(4-((E)-2-(2-chloro-4-fluorophenyl)-1-(1-(tetrahydro-2H-pyran-2-yl)-1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylate
  • Step 3 (E)-Ethyl 3-(4-((E)-2-(2-chloro-4-fluorophenyl)-1-(1H-indazol-5-yl)but-1-en-1-yl)phenypacrylate hydrochloride
  • Step 1 (E)-4-(2-(2,4-Dichlorophenyl)-1-(1-(tetrahydro-2H-pyran-2-yl)-1H-indazol-5-yl)but-1-en-1-yl)benzaldehyde
  • Step 2 (E)-Ethyl 3-(4-((E)-2-(2,4-dichlorophenyl)-1-(1-(tetrahydro-2H-pyran-2-yl)-1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylate
  • Step 3 (E)-Ethyl 3-(4-((E)-2-(2,4-dichlorophenyl)-1-(1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylate
  • N-Iodosuccinimide (2.5 g, 11 mmol) was added to a suspension of 4-(E-3-ethoxy-3-oxo-1-propen-1-yl)phenylboronic acid (2.2 g, 10 mmol) in CH 3 CN (50 mL) at room temperature. The reaction was covered with foil, stirred for ⁇ 26 hours, and then diluted with EtOAc. The resulting mixture was washed with water (2 ⁇ 100 mL), washed with sodium thiosulfate (100 mL), dried (MgSO 4 ), filtered, and concentrated.
  • Step 2 (E)-Ethyl 3-(44(Z)-1-(1-(tetrahydro-2H-pyran-2-yl)-1H-indazol-5-yl)-2-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)but-1-en-1-yl)phenyl)acrylate
  • Step 1 (E)-Ethyl 3-(4-((E)-2-cyclobutyl-1-(1-methyl-1H-indazol-5-yl)-2-phenylvinyl)phenyl)acrylate
  • Step 2 (E)-3-(4-0E)-2-Cyclobutyl-1-(1-methyl-1H-indazol-5-yl)-2-phenylvinyl)phenyl)acrylic acid
  • MCF7 cells were maintained in RPMI 1640 supplemented with 10% FCS. Transcriptional assays were performed by seeding 100 ⁇ L of cells at a density of 250,000 cells/mL into 96-well cell culture plates in RPMI 1640 supplemented with 10% charcoal stripped serum and allowed to attach overnight. Cells were transiently transfected using Lipofectin (Life Technologies) according to the manufacturer's protocol. Triplicate transfections were performed using 300 ng 3 ⁇ ERE-TK-Luc (reporter vector), 50 ng CMVpRL (normalization vector), and 130 ng pCMX (filler DNA). Transfected cells were incubated overnight then treated with ligand.
  • the compounds were serially diluted and 50 ⁇ L of compound plus RPMI 1640 supplemented with charcoal stripped serum was added to the cells.
  • the compounds were serially diluted and 50 ⁇ L of compound with RPMI plus 17 ⁇ -estradiol supplemented with charcoal stripped serum were added to the cells. The final 17 ⁇ -estradiol concentration used in the antagonist assays was 0.1 nM. Following 24 hour incubation the medium was removed and the cells were lysed in 40 ⁇ L of lysis buffer (25 mM Tris Phosphate, 2 mM CDTA, 10% Glycerol, 0.5% Triton X-100, 2 mM DTT).
  • Firefly luciferase activity was measured immediately following the addition of 40 ⁇ L luciferase buffer (20 mM tricine, 0.1 mM EDTA, 1.07 mM (MgCO 3 ) 4 Mg(OH) 2 .5H 2 O, 2.67 mM MgSO 4 , 33.3 mM DTT, 270 ⁇ M Coenzyme A, 470 ⁇ M luciferin, 530 ⁇ M ATP).
  • Renilla luciferase was measured following the addition of 40 ⁇ L colelenterazine buffer (1.1 M NaCl, 2.2 mM Na 2 EDTA, 0.22 M KxPO 4 (pH 5.1), 0.44 mg/mL BSA, 1.3 mM NaN 3 , 1.43 ⁇ M coelenterazine, final pH adjusted to 5.0).
  • MCF-7 cells were adjusted to a concentration of 20,000 cells per mL in RPMI containing 10% FBS and 20 mM HEPES. 16 microliters of the cell suspension (320 cells) was added to each well of a 384 well plate, and the cells were incubated overnight to allow the cells to adhere. The following day an eleven point, serial semilog dilution of each compound was added to the cells in 16 ⁇ L at a final concentration ranging from 0.3-0.000003 ⁇ M.
  • Viability effects in additional ER+ breast cancer cell lines including BT474, CAMA1, MDA-MB-361, ZR-75-1, T47D, can be profiled in assays similar to Example 44.
  • MCF-7 cells were adjusted to a concentration of 200,000 cells per mL in RPMI containing 10% charcoal-stripped FBS and 20 mM HEPES. 16 microliters of the cell suspension (3200 cells) was added to each well of a poly-D-lysine 384 well plate, and the cells were incubated overnight to allow the cells to adhere. The following day an eleven point, serial semilog dilution of each compound was added to the cells in 16 ⁇ L at a final concentration ranging from 0.3-0.000003 ⁇ M. At 4 or 24 hr post compound addition, the cells were fixed (10% formalin in PBS) for 20 minutes.
  • LICOR blocking buffer 50 ⁇ l/well, 90′.
  • the wells were then incubated overnight at 4° C. with SP1 rabbit monoclonal Ab (Thermo Scientific) diluted 1:1000 in LICOR blocking buffer/0.1% Tween-20.
  • Wells which were treated with blocking buffer with Tween but no antibody were used as a background control.
  • Wells were washed with 0.1% Tween-20/PBS and then incubated in goat anti-rabbit IRDyeTM 800CW (LICOR Inc.; 1:1000) and DRAQ5 DNA dye (1:2000 for 2 mM stock) diluted in LICOR blocking buffer containing 0.1% Tween-20 and 0.01% SDS for 60 minutes.
  • Subconfuent Ishikawa cells in a T225 are incubated 24 hours in an estrogen free basal medium (EFBM) consisting of DMEM:Ham's F-12 50:50 phenol red free basal medium containing 5% Charcoal Dextran treated FBS and 20 mM HEPES.
  • EFBM estrogen free basal medium
  • Cells are plated the following day in EFBM in clear 384 well plates at a concentration of 2.5 ⁇ 105 cells per mL, 16 ⁇ l per well (4000 cells per well).
  • a 12 point semilog dilution of each compound is carried out in DMSO and subsequently diluted in EFBM.
  • An equal volume of compound in EFBM is added immediately after plating cells, and the cells are incubated for 3 days.
  • the cells are fixed with 5% formalin, and rinsed with PBS.
  • Alkaline Phosphatase substrate 4-Nitrophenyl phosphate disodium salt hexahydrate is added to a solution containing 2 mM MgCl 2 , 1 M diethanolamine, and adjusted to pH 9.0.
  • the substrate solution is added to the cell cultures (16 ⁇ L per well), and OD405 is measured in a multiwall plate spectrophotometer when the optical density at 405 nm wavelength of cells treated with 17 ⁇ -estradiol in the concentration range of 1-30 nM reaches 1.0-1.2 absorbance units.
  • BG-1 BG-1 cells were adjusted to a concentration of 20,000 cells per mL in RPMI containing 10% FBS and 20 mM HEPES. 16 microliters of the cell suspension (320 cells) was added to each well of a 384 well plate, and the cells were incubated overnight to allow the cells to adhere. The following day an eleven point, serial semilog dilution of each compound was added to the cells in 16 ⁇ L at a final concentration ranging from 0.3-0.000003 ⁇ M.
  • Viability effects in additional ER+ ovarian cancer cell lines, including A1847, SKOV3, SW626, A2780, can be profiled in assays similar to Example 47.
  • BG-1 cells were adjusted to a concentration of 200,000 cells per mL in RPMI containing 10% charcoal-stripped FBS and 20 mM HEPES. 16 microliters of the cell suspension (3200 cells) was added to each well of a poly-D-lysine 384 well plate, and the cells were incubated overnight to allow the cells to adhere. The following day an eleven point, serial semilog dilution of each compound was added to the cells in 16 ⁇ L at a final concentration ranging from 0.3-0.000003 ⁇ M. At 4 or 24 hr post compound addition, the cells were fixed (10% formalin in PBS) for 20 minutes.
  • LICOR blocking buffer 50 ⁇ l/well, 90′. The wells were then incubated overnight at 4° C. with ER1D5 (Santa Cruz Biotechnology) diluted 1:100 in LICOR blocking buffer/0.1% Tween-20. Wells which were treated with blocking buffer with Tween but no antibody were used as a background control. Wells were washed with 0.1% Tween-20/PBS and then incubated in goat anti-mouse IRDyeTM 800CW (LICOR Inc.; 1:1000) and DRAQ5 DNA dye (1:2000 for 2 mM stock) diluted in LICOR blocking buffer containing 0.1% Tween-20 and 0.01% SDS for 60 minutes.
  • ER1D5 Santa Cruz Biotechnology
  • cancer cell lines contemplated for testing compounds described herein include: ER-positive endometrial cell lines (Ishikawa, ECC1, HEC-1, EnCa-101) and ER-positive cervical cell lines (Caski, HeLa, SiHa).
  • Time release pellets containing 0.72 mg 17- ⁇ Estradiol were subcutaneously implanted into nu/nu mice.
  • MCF-7 cells were grown in RPMI containing 10% FBS at 5% CO 2 , 37° C. Cells were spun down and re-suspended in 50% RPMI (serum free) and 50% Matrigel at 1 ⁇ 10 7 cells/mL. MCF-7 cells were subcutaneously injected (100 ⁇ L/animal) on the right flank 2-3 days post pellet implantation. Tumor volume (length ⁇ width 2 /2) was monitored bi-weekly. When tumors reached an average volume of ⁇ 200 mm 3 animals were randomized and treatment was started. Animals were treated with Vehicle or Compound daily for 4 weeks. Tumor volume and body weight were monitored bi-weekly throughout the study. At the conclusion of the treatment period, plasma and tumor samples were taken for pharmacokinetic and pharmacodynamic analyses, respectively.
  • mice Female nu/nu mice (with supplemental 17-(Estradiol pellets; 0.72 mg; 60 day slow release) bearing MCF-7 tumors (mean tumor volume 200 mm 3 ) were treated with Tamoxifen (citrate) by oral gavage. Tumor volume (length ⁇ width 2 /2) and body weight were monitored twice weekly. Following a significant anti-tumor response in which tumor volume remained static, evident tumor growth was first observed at approximately 100 days of treatment. At 120 days of treatment, tamoxifen dose was increased. Rapidly growing tumors were deemed tamoxifen resistant and selected for in vivo passage into new host animals.
  • Tumor Fragments ( ⁇ 100 mm 3 /animal) from the tamoxifen resistant tumors were subcutaneously implanted into the right flank of female nu/nu mice (with 17-0 Estradiol pellets (0.72 mg; 60 day slow release)). Passaged tumors were maintained under constant Tamoxifen selection, and Tumor volume (length ⁇ width 2 /2) was monitored weekly. When tumor volume reached ⁇ 150-250 mm 3 , animals were randomized into treatment groups (mean tumor volume 200 mm 3 ) and tamoxifen treatment was terminated (except for a tamoxifen control arm) Animals were treated with Vehicle or Compound daily for 4 weeks. Tumor volume and body weight were monitored twice weekly for the duration of the study. At the conclusion of the treatment period; plasma and tumor samples were taken for pharmacokinetic and pharmacodynamic analyses, respectively.
  • Time release pellets (0.72 mg 17-0 Estradiol/60 days) were subcutaneously implanted into female nu/nu mice.
  • BG-1 cells were grown in DMEM Ham's F-12 50/50 containing 10% FBS, 10 mM Sodium Pyruvate, 10 mM Non-Essential Amino Acids at 5% CO 2 , 37° C. Cells were spun down and re-suspended in 50% DMEM Ham's F-12 (serum free) and 50% Matrigel at 5 ⁇ 10 7 cells/mL.
  • BG-1 cells were subcutaneously injected (100 ⁇ L/animal) on the right flank 2-3 days post pellet implantation. Tumor volume (length ⁇ width 2 /2) was monitored bi-weekly.
  • tumors reached an average volume of ⁇ 250 mm 3 animals were randomized and treatment was started. Animals were treated with Vehicle or Compound daily for 4 weeks. Tumor volume and body weight were monitored bi-weekly throughout the study. At the conclusion of the treatment period; plasma and tumor samples were taken for pharmacokinetic and pharmacodynamic analyses, respectively.
  • the purpose of this study is to assess the efficacy of treatment of metastatic breast cancer with a compound of Formula (I), or a pharmaceutically acceptable salt thereof, collect information on any side effects the compound of Formula (I), or a pharmaceutically acceptable salt thereof, may cause, and evaluate the pharmacokinetic properties of the compound of Formula (I), or a pharmaceutically acceptable salt thereof.
  • Intervention Patients are administered 1-50 mg/kg of a compound of Formula (I), or a pharmaceutically acceptable salt thereof, per day or twice a day.
  • the primary outcome measure is the patient's tumor response and/or disease control to a compound of Formula (I), or a pharmaceutically acceptable salt thereof, as first- and/or second-line treatment in patients with estrogen receptor (ER) positive metastatic breast cancer.
  • ER estrogen receptor
  • the second outcome measures are (a) an evaluation of the side-effects of a compound of Formula (I), or a pharmaceutically acceptable salt thereof; (b) an evaluation of the pharmacokinetic properties of a compound of Formula (I), or a pharmaceutically acceptable salt thereof; (c) an evaluation of the proportion of patients that have complete or partial response or stable disease at defined timepoint; (d) an evaluation of the time to progression and overall survival of patients treated with a compound of Formula (I), or a pharmaceutically acceptable salt thereof; and (e) biomarkers predictive of clinical response.
  • Patients will be given a compound of Formula (I), or a pharmaceutically acceptable salt thereof, orally once or twice a day. Prior to each dosing cycle of a compound of Formula (I), or a pharmaceutically acceptable salt thereof, a physical exam, blood work and assessment of any side effects will be performed. Every 12 weeks the patient's cancer will be re-evaluated with either a CT scan or MRI to determine whether the treatment is working. Participation in this study will last until disease progression or unacceptable toxicity.
  • Eligibility Female subjects, 18 years and older.
  • Inclusion Criteria Histologically or cytologically confirmed diagnosis of invasive breast cancer, Stage IV disease. At least one measurable target lesion as defined by RECIST that has not been previously treated with local therapy. Post-menopausal status. ER positive breast cancer. HER2-negative breast cancer. Up to one prior hormonal therapy for advanced or metastatic disease. ECOG performance status 0-1. Life expectancy>12 weeks. Adequate liver and bone marrow function: AST ⁇ 2.5 ⁇ ULN; Bilirubin ⁇ 1.5 ⁇ ULN; ANC>1,500/ul; platelet count>100,000/ul; normal PT and PTT. At least 2 weeks since prior radiation and recovered from treatment-related toxicity.
  • HER2-positive breast cancer Prior chemotherapy regimen for metastatic disease. History of, or presence of brain metastases. Concurrent investigational drug treatment. Prior bone marrow or stem cell transplant. History of other malignancy within the last 5 years, not including curatively-treated carcinoma in situ of the cervix or non-melanoma skin cancer. Uncontrolled infection. Active bleeding, or history of bleeding requiring transfusion.
  • a parenteral pharmaceutical composition suitable for administration by injection 100 mg of a compound of Formula (I), or a water-soluble salt of a compound of Formula (I), is dissolved in sterile water and then mixed with 10 mL of 0.9% sterile saline. The mixture is incorporated into a dosage unit form suitable for administration by injection
  • the following ingredients are mixed to form an injectable formulation: 1.2 g of a compound of Formula (I), or a pharmaceutically acceptable salt thereof, 2.0 mL of sodium acetate buffer solution (0.4 M), HCl (1 N) or NaOH (1 M) (q.s. to suitable pH), water (distilled, sterile) (q.s. to 20 mL). All of the above ingredients, except water, are combined and stirred and if necessary, with slight heating if necessary. A sufficient quantity of water is then added.
  • an aqueous 20% propylene glycol solution is prepared. To this is added a sufficient amount of compound of Formula (I), or a pharmaceutically acceptable salt thereof, to provide a 20 mg/mL solution.
  • a pharmaceutical composition for oral delivery 100-500 mg of a compound of Formula (I), or a pharmaceutically acceptable salt thereof, is mixed with starch. The mixture is incorporated into an oral dosage unit such as a hard gelatin capsule, which is suitable for oral administration. In another embodiment, 100-500 mg of a compound of Formula (I), or a pharmaceutically acceptable salt thereof, is placed into Size 4 capsule, or size 1 capsule (hypromellose or hard gelatin) and the capsule is closed.
  • a tablet is prepared by mixing 48% by weigh of a compound of Formula (I), or a pharmaceutically acceptable salt thereof, 45% by weight of microcrystalline cellulose, 5% by weight of low-substituted hydroxypropyl cellulose, and 2% by weight of magnesium stearate. Tablets are prepared by direct compression. The total weight of the compressed tablets is maintained at 250-500 mg.
  • a compound of Formula (I), or a pharmaceutically acceptable salt thereof is mixed with hydroxypropyl celluose, propylene glycol, isopropyl myristate and purified alcohol USP.
  • the resulting gel mixture is then incorporated into containers, such as tubes, which are suitable for topical administration.

Abstract

Described herein are compounds that are estrogen receptor modulators. Also described are pharmaceutical compositions and medicaments that include the compounds described herein, as well as methods of using such estrogen receptor modulators, alone and in combination with other compounds, for treating diseases or conditions that are mediated or dependent upon estrogen receptors.

Description

    RELATED APPLICATIONS
  • This application claims the benefit of U.S. provisional patent application No. 61/383,659 entitled “ESTROGEN RECEPTOR MODULATORS AND USES THEREOF” filed on Sep. 16, 2010; U.S. provisional patent application No. 61/410,727 entitled “ESTROGEN RECEPTOR MODULATORS AND USES THEREOF” filed on Nov. 5, 2010; U.S. provisional patent application No. 61/446,967 entitled “ESTROGEN RECEPTOR MODULATORS AND USES THEREOF” filed on Feb. 25, 2011; United Kingdom Patent Application No 11 04288.4 entitled “ESTROGEN RECEPTOR MODULATORS AND USES THEREOF” filed on Mar. 15, 2011; each of which is incorporated by reference in its entirety.
  • FIELD OF THE INVENTION
  • Described herein are compounds, including pharmaceutically acceptable salts, solvates, metabolites, prodrugs thereof, methods of making such compounds, pharmaceutical compositions comprising such compounds, and methods of using such compounds to treat, prevent or diagnose diseases or conditions that are estrogen sensitive, estrogen receptor dependent or estrogen receptor mediated.
  • BACKGROUND OF THE INVENTION
  • The estrogen receptor (“ER”) is a ligand-activated transcriptional regulatory protein that mediates induction of a variety of biological effects through its interaction with endogenous estrogens. Endogenous estrogens include 17β-estradiol and estrones. ER has been found to have two isoforms, ER-α and ER-β. Estrogens and estrogen receptors are implicated in a number of diseases or conditions, such as breast cancer, ovarian cancer, colon cancer, prostate cancer, endometrial cancer, uterine cancer, as well as others diseases or conditions.
  • SUMMARY OF THE INVENTION
  • In one aspect, presented herein are compounds of Formula (I), or a pharmaceutically acceptable salt, or N-oxide thereof, that diminish the effects of estrogens with estrogen receptors and/or lower the concentrations of estrogen receptors, and therefore, are useful as agents for the treatment or prevention of diseases or conditions in which the actions of estrogens and/or estrogen receptors are involved in the etiology or pathology of the disease or condition or contribute to at least one symptom of the disease or condition and wherein such actions of estrogens and/or estrogen receptors are undesirable. In some embodiments, compounds disclosed herein are estrogen receptor degrader compounds.
  • In one aspect, the compound of Formula (I), or a pharmaceutically acceptable salt, or N-oxide thereof, is useful for the treatment of ER-related diseases or conditions including, but not limited to, ER-a dysfunction associated with cancer (e.g. bone cancer, breast cancer, colorectal cancer, endometrial cancer, prostate cancer, ovarian and uterine cancer), leiomyoma (e.g. uterine leiomyoma), central nervous system (CNS) defects (e.g. alcoholism, migraine), cardiovascular system defects (e.g. aortic aneurysm, susceptibility to myocardial infarction, aortic valve sclerosis, cardiovascular disease, coronary artery disease, hypertension), hematological system defects (e.g. deep vein thrombosis), immune and inflammation diseases (e.g. Graves' Disease, arthritis, multiple sclerosis, cirrhosis), susceptibility to infection (e.g. hepatitis B, chronic liver disease), metabolic defects (e.g. bone density, cholestasis, hypospadias, obesity, osteoarthritis, osteopenia, osteoporosis), neurological defects (e.g. Alzheimer's disease, Parkinson's disease, migraine, vertigo), psychiatric defects (e.g. anorexia nervosa, attention deficity hyperactivity disorder (ADHD), dementia, major depressive disorder, psychosis) and reproductive defects (e.g. age of menarche, endometriosis, infertility).
  • In one aspect, described herein are compounds of Formula (I), pharmaceutically acceptable salts, solvates, N-oxides, metabolites and prodrugs thereof. Compounds of Formula (I) are estrogen receptor modulators. In some embodiments, the compound of Formula (I) is an estrogen receptor antagonist. In some embodiments, the compound of Formula (I) is an estrogen receptor degrader. In some embodiments, the compound of Formula (I) is an estrogen receptor antagonist as well as an estrogen receptor degrader. In some embodiments, the compound of Formula (I) displays minimal or no estrogen receptor agonist activity. In some embodiments, in the context of treating cancers, the compound of Formula (I) offers improved therapeutic activity characterized by complete or longer-lasting tumor regression, a lower incidence or rate of development of resistance to treatment, and/or a reduction in tumor invasiveness.
  • In one aspect, provided herein is a compound of Formula (I), or a pharmaceutically acceptable salt, or N-oxide thereof:
  • Figure US20120071535A1-20120322-C00001
      • wherein,
      • Z is —OH or —OR10;
      • R2 is C1-C4alkyl, C1-C4-fluoroalkyl, C1-C4deuteroalkyl, C3-C6cycloalkyl, or —C1-C4alkylene-W;
        • W is hydroxy, halogen, CN, C1-C4alkyl, C1-C4haloalkyl, C1-C4alkoxy, C1-C4haloalkoxy, or C3-C6cycloalkyl;
      • each R3 is independently halogen, C1-C4alkyl, or C1-C4-fluoroalkyl;
      • each R4 is independently halogen, —CN, —OR9, —S(═O)2R19, C1-C4alkyl, C1-C4-fluoroalkyl, or C1-C4heteroalkyl;
      • each R5 is independently halogen, —CN, —OR9, —S(═O)2R19, C1-C4alkyl, C1-C4-fluoroalkyl, or C1-C4heteroalkyl;
      • R6 is H, C1-C4alkyl, or halogen; R7 is H, C1-C4alkyl, or halogen;
      • R9 is H, C1-C6alkyl, C1-C6-fluoroalkyl, or C3-C6cycloalkyl;
      • R10 is C1-C6alkyl;
      • m is 0, 1, or 2; n is 0, 1, 2, 3, or 4; and p is 0, 1, or 2.
  • In some embodiments, the compound of Formula (I), or a pharmaceutically acceptable salt thereof, or a pharmaceutically acceptable salt, or N-oxide thereof, has the structure of Formula (II), or a pharmaceutically acceptable salt, or N-oxide thereof.
  • In some embodiments, the compound of Formula (I), or a pharmaceutically acceptable salt thereof, is a compound described in Table 1, or a pharmaceutically acceptable salt thereof.
  • Throughout the specification, groups and substituents thereof can be chosen by one skilled in the field to provide stable moieties and compounds.
  • Compounds disclosed herein are estrogen receptor modulators. In some embodiments, compounds disclosed herein have high specificity for the estrogen receptor and have desirable, tissue-selective pharmacological activities. Desirable, tissue-selective pharmacological activities include, but are not limited to, ER antagonist activity in breast cells and minimal or no ER agonist activity in uterine cells. In some embodiments, compounds disclosed herein are estrogen receptor degraders that display full estrogen receptor antagonist activity with negligible or minimal estrogen receptor agonist activity.
  • In some embodiments, compounds disclosed herein are estrogen receptor degraders. In some embodiments, compounds disclosed herein are estrogen receptor antagonists. In some embodiments, compounds disclosed herein have minimal or negligible estrogen receptor agonist activity.
  • In some embodiments, presented herein are compounds selected from active metabolites, tautomers, pharmaceutically acceptable solvates, pharmaceutically acceptable salts or prodrugs of a compound of Formula (I).
  • Also described are pharmaceutical compositions comprising a therapeutically effective amount of a compound of Formula (I), or a pharmaceutically acceptable salt, or N-oxide thereof. In some embodiments, the pharmaceutical composition also contains at least one pharmaceutically acceptable inactive ingredient. In some embodiments, the pharmaceutical composition is formulated for intravenous injection, subcutaneous injection, oral administration, or topical administration. In some embodiments, the pharmaceutical composition is a tablet, a pill, a capsule, a liquid, a suspension, a gel, a dispersion, a suspension, a solution, an emulsion, an ointment, or a lotion.
  • In some embodiments, the pharmaceutical composition further comprises one or more additional therapeutically active agents selected from: corticosteroids, anti-emetic agents, analgesics, anti-cancer agents, anti-inflammatories, kinase inhibitors, antibodies, HSP90 inhibitors, histone deacetylase (HDAC) inhibitors, poly ADP-ribose polymerase (PARP) inhibitors, and aromatase inhibitors.
  • In some embodiments, provided herein is a method comprising administering a compound of Formula (I), or a pharmaceutically acceptable salt, or N-oxide thereof to a human with a diseases or condition that is estrogen sensitive, estrogen receptor meditated or estrogen receptor dependent. In some embodiments, the human is already being administered one or more additional therapeutically active agents other than a compound of Formula (I), or a pharmaceutically acceptable salt, or N-oxide thereof. In some embodiments, the method further comprises administering one or more additional therapeutically active agents other than a compound of Formula (I), or a pharmaceutically acceptable salt, or N-oxide thereof.
  • In some embodiments, the one or more additional therapeutically active agents other than a compound of Formula (I), or a pharmaceutically acceptable salt, or N-oxide thereof are selected from: corticosteroids, anti-emetic agents, analgesics, anti-cancer agents, anti-inflammatories, kinase inhibitors, antibodies, HSP90 inhibitors, histone deacetylase (HDAC) inhibitors, and aromatase inhibitors.
  • In some embodiments, the methods further comprise administering to the mammal radiation therapy. In certain embodiments, the compound of the methods is administered prior to or following surgery. In certain embodiments, the methods comprise administering to the mammal at least one additional anti-cancer agent.
  • In some embodiments, there are provided methods of treating cancer in a mammal comprising administering to the mammal a therapeutically effective amount of a compound of Formula (I), or a pharmaceutically acceptable salt, or N-oxide thereof. In certain embodiments, the cancer is breast cancer, ovarian cancer, endometrial cancer, prostate cancer, uterine cancer, cervical cancer or lung cancer. In certain embodiments, the cancer is breast cancer. In certain embodiments, the cancer is a hormone dependent cancer. In certain embodiments, the cancer is an estrogen receptor dependent cancer. In certain embodiments, the cancer is an estrogen-sensitive cancer. In certain embodiments, the cancer is resistant to anti-hormonal treatment. In certain embodiments, the cancer is an estrogen-sensitive cancer or an estrogen receptor dependent cancer that is resistant to anti-hormonal treatment. In some embodiments, the anti-hormonal treatment includes treatment with at least one agent selected from tamoxifen, fulvestrant, steroidal aromatase inhibitors, and non-steroidal aromatase inhibitors. In some embodiments, the methods further comprise administering to the mammal radiation therapy. In certain embodiments, the compound of the methods is administered prior to or following surgery. In certain embodiments, the methods comprise administering to the mammal at least one additional anti-cancer agent.
  • In some embodiments, there are provided methods of treating hormone receptor positive metastatic breast cancer in a postmenopausal woman with disease progression following anti-estrogen therapy comprising administering to the woman an estrogen receptor degrading compound of Formula (I), or a pharmaceutically acceptable salt, or N-oxide thereof. In some embodiments, the methods further comprise administering to the mammal radiation therapy. In certain embodiments, the compound of the methods is administered prior to or following surgery. In certain embodiments, the methods comprise administering to the mammal at least one additional anti-cancer agent.
  • In some embodiments, there are provided methods of treating a hormonal dependent benign or malignant disease of the breast or reproductive tract in a mammal comprising administering to the mammal an effective amount of a compound of Formula (I), a pharmaceutically acceptable salt, or N-oxide thereof. In certain embodiments, the benign or malignant disease is breast cancer. In some embodiments, the methods further comprise administering to the mammal radiation therapy. In certain embodiments, the compound of the methods is administered prior to or following surgery. In certain embodiments, the methods comprise administering to the mammal at least one additional anti-cancer agent.
  • In some embodiments, a compound of Formula (I), or a pharmaceutically acceptable salt, or N-oxide thereof is used to treat hormone receptor positive metastatic breast cancer in a postmenopausal woman with disease progression following anti-estrogen therapy. In some embodiments, the compound is used to treat a hormonal dependent benign or malignant disease of the breast or reproductive tract in a mammal.
  • Also provided is a method of reducing ER activation in a mammal comprising administering to the mammal at least one compound having the structure of Formula (I), or a pharmaceutically acceptable salt, or N-oxide thereof. In some embodiments, the method comprises reducing ER activation in breast cells, ovarian cells, colon cells, prostate cells, endometrial cells, or uterine cells in the mammal. In some embodiments, the method of reducing ER activation in the mammal comprises reducing the binding of estrogens to estrogen receptors in the mammal In some embodiments, the method of reducing ER activation in the mammal comprises reducing ER concentrations in the mammal.
  • In one aspect is the use of a compound of Formula (I), or a pharmaceutically acceptable salt, or N-oxide thereof in the manufacture of a medicament for the treatment of diseases or conditions that are estrogen sensitive, estrogen receptor dependent or estrogen receptor mediated. In some embodiments, the disease or condition is breast cancer, ovarian cancer, colon cancer, prostate cancer, endometrial cancer, or uterine cancer. In some embodiments, the disease or condition is bone cancer, breast cancer, colorectal cancer, endometrial cancer, prostate cancer, ovarian cancer, uterine cancer, cervical cancer, lung cancer, alcoholism, migraine, aortic aneurysm, susceptibility to myocardial infarction, aortic valve sclerosis, cardiovascular disease, coronary artery disease, hypertension, deep vein thrombosis, Graves' Disease, arthritis, multiple sclerosis, cirrhosis, hepatitis B, chronic liver disease, bone density, cholestasis, hypospadias, obesity, osteoarthritis, osteopenia, osteoporosis, Alzheimer's disease, Parkinson's disease, migraine, vertigo, anorexia nervosa, attention deficit hyperactivity disorder (ADHD), dementia, major depressive disorder, psychosis, age of menarche, endometriosis, or infertility in a mammal. In some embodiments, the disease or condition is described herein.
  • In some cases disclosed herein is the use of a compound of Formula (I), or a pharmaceutically acceptable salt, or N-oxide thereof in the treatment or prevention of diseases or conditions that are estrogen sensitive, estrogen receptor dependent or estrogen receptor mediated. In some embodiments, the disease or condition is described herein.
  • In any of the embodiments disclosed herein, the mammal is a human.
  • In some embodiments, compounds provided herein are used to diminish, reduce, or eliminate the activity of estrogen receptors.
  • Other objects, features and advantages of the compounds, methods and compositions described herein will become apparent from the following detailed description. It should be understood, however, that the detailed description and the specific examples, while indicating specific embodiments, are given by way of illustration only, since various changes and modifications within the spirit and scope of the instant disclosure will become apparent to those skilled in the art from this detailed description.
  • DETAILED DESCRIPTION OF THE INVENTION
  • Estrogen receptor alpha (ER-α; NR3A1) and estrogen receptor beta (ER-β; NR3A2) are steroid hormone receptors, which are members of the large nuclear receptor superfamily. Nuclear receptors share a common modular structure, which minimally includes a DNA binding domain (DBD) and a ligand binding domain (LBD). Steroid hormone receptors are soluble, intracellular proteins that act as ligand-regulated transcription factors. Vertebrates contain five closely related steroid hormone receptors (estrogen receptor, androgen receptor, progesterone receptor, glucocorticoid receptor, mineralcorticoid receptor), which regulate a wide spectrum of reproductive, metabolic and developmental activities. The activities of ER are controlled by the binding of endogenous estrogens, including 17β-estradiol and estrones.
  • The ER-α gene is located on 6q25.1 and encodes a 595 AA protein. The ER-β gene resides on chromosome 14q23.3 and produces a 530 AA protein. However, due to alternative splicing and translation start sites, each of these genes can give rise to multiple isoforms. In addition to the DNA binding domain (called C domain) and ligand binding domain (E domain) these receptors contain an N-terminal (A/B) domain, a hinge (D) domain that links the C and E domains, and a C-terminal extension (F domain) (Gronemeyer and Laudet; Protein Profile 2: 1173-1308, 1995). While the C and E domains of ER-α and ER-β are quite conserved (95% and 55% amino acid identity, respectively), conservation of the A/B, D and F domains is poor (below 30% amino acid identity). Both receptors are involved in the regulation and development of the female reproductive tract but also play various roles in the central nervous system, cardiovascular systems and bone metabolism.
  • The ligand binding pocket of steroid hormone receptors is deeply buried within the ligand binding domain. Upon binding, the ligand becomes part of the hydrophobic core of this domain. Consequently most steroid hormone receptors are instable in the absence of hormone and require assistance from chaperones, such as Hsp90, in order to maintain hormone-binding competency. The interaction with Hsp90 also controls nuclear translocation of these receptors. Ligand-binding stabilizes the receptor and initiates sequential conformational changes that release the chaperones, alter the interactions between the various receptor domains and remodel protein interaction surfaces that allow these receptors to translocate into the nucleus, bind DNA and engage in interactions with chromatin remodeling complexes and the transcriptional machinery. Although ER can interact with Hsp90, this interaction is not required for hormone binding and, dependent on the cellular context, apo-ER can be both cytoplasmic and nuclear. Biophysical studies indicated that DNA binding rather than ligand binding contributes to the stability of the receptor (Greenfield et al., Biochemistry 40: 6646-6652, 2001).
  • ER can interact with DNA either directly by binding to a specific DNA sequence motif called estrogen response element (ERE) (classical pathway), or indirectly via protein-protein interactions (nonclassical pathway) (Welboren et al., Endocrine-Related Cancer 16: 1073-1089, 2009). In the nonclassical pathway, ER has been shown to tether to other transcription factors including SP-1, AP-1 and NF-κB. These interactions appear to play critical roles in the ability of ER to regulate cell proliferation and differentiation.
  • Both types of ER DNA interactions can result in gene activation or repression dependent on the transcriptional coregulators that are recruited by the respective ER-ERE complex (Klinge, Steroid 65: 227-251, 2000). The recruitment of coregulators is primarily mediated by two protein interaction surfaces, the AF2 and AF1. AF2 is located in the ER E-domain and its conformation is directly regulated by the ligand (Brzozowski et al., Nature 389: 753-758, 1997). Full agonists appear to promote the recruitment of co-activators, whereas weak agonists and antagonists facilitate the binding of co-repressors. The regulation of protein with the AF 1 is less well understood but can be controlled by serine phosphorylation (Ward and Weigel, Biofactors 35: 528-536, 2009). One of the involved phosphorylation sites (S118) appears to control the transcriptional activity of ER in the presence of antagonists such as tamoxifen, which plays an important role in the treatment of breast cancer. While full agonists appear to arrest ER in certain conformation, weak agonists tend to maintain ER in equilibrium between different conformations, allowing cell-dependent differences in co-regulator repertoires to modulate the activity of ER in a cell-dependent manner (Tamrazi et al., Mol. Endocrinol. 17: 2593-2602, 2003). Interactions of ER with DNA are dynamic and include, but are not limited to, the degradation of ER by the proteasome (Reid et al., Mol Cell 11: 695-707, 2003). The degradation of ER with ligands provides an attractive treatment strategy for disease or conditions that estrogen-sensitive and/or resistant to available anti-hormonal treatments.
  • ER signaling is crucial for the development and maintenance of female reproductive organs including breasts, ovulation and thickening of the endometrium. ER signaling also has a role in bone mass, lipid metabolism, cancers, etc. About 70% of breast cancers express ER-α (ER-α positive) and are dependent on estrogens for growth and survival. Other cancers also are thought to be dependent on ER-α signaling for growth and survival, such as for example ovarian and endometrial cancers. The ER-α antagonist tamoxifen has been used to treat early and advanced ER-α positive breast cancer in both pre- and post-menopausal women. Fulvestrant (Faslodex™) a steroid-based ER antagonist is used to treat breast cancer in women which has have progressed despite therapy with tamoxifen. Steroidal and non-steroidal aromatase inhibitors are also used to treat cancers in humans. In some embodiments, the steroidal and non-steroidal aromatase inhibitors block the production of estrogen from androstenedione and testosterone in post-menopausal women, thereby blocking ER dependent growth in the cancers. In addition to these anti-hormonal agents, progressive ER positive breast cancer is treated in some cases with a variety of other chemotherapeutics, such as for example, the anthracylines, platins, taxanes. In some cases, ER positive breast cancers that harbor genetic amplication of the ERB-B/HER2 tyrosine kinase receptor are treated with the monoclonal antibody trastuzumab (Herceptin™) or the small molecule pan-ERB-B inhibitor lapatinib. Despite this battery of anti-hormonal, chemotherapeutic and small-molecule and antibody-based targeted therapies, many women with ER-α positive breast develop progressive metastatic disease and are in need of new therapies. Importantly, the majority of ER positive tumors that progress on existing anti-hormonal, as well as and other therapies, are thought to remain dependent on ER-α for growth and survival. Thus, there is a need for new ER-α targeting agents that have activity in the setting of metastatic disease and acquired resistance.
  • In one aspect, described herein are compounds that are selective estrogen receptor modulators (SERMs). In specific embodiments, the SERMs described herein are selective estrogen receptor degraders (SERDs). In some embodiments, in cell-based assays the compounds described herein result in a reduction in steady state ER-α levels (i.e. ER degradation) and are useful in the treatment of estrogen sensitive diseases or conditions and/or diseases or conditions that have developed resistant to anti-hormonal therapies. In some embodiments, compounds disclosed herein minimize levels of the estrogen receptor in the nucleus.
  • Given the central role of ER-α in breast cancer development and progression, compounds disclosed herein are useful in the treatment of breast cancer, either alone or in combination with other agent agents that modulate other critical pathways in breast cancer, including but not limited to those that target IGF1R, EGFR, erB-B2 and 3 the PI3K/AKT/mTOR axis, HSP90, PARP or histone deacetylases.
  • Given the central role of ER-α in breast cancer development and progression, compounds disclosed herein are useful in the treatment of breast cancer, either alone or in combination with other agent used to treat breast cancer, including but not limited to aromatase inhibitors, anthracylines, platins, nitrogen mustard alkylating agents, taxanes. Illustrative agent used to treat breast cancer, include, but are not limited to, paclitaxel, anastrozole, exemestane, cyclophosphamide, epirubicin, fulvestrant, letrozole, gemcitabine, trastuzumab, pegfilgrastim, filgrastim, tamoxifen, docetaxel, toremifene, vinorelbine, capecitabine, ixabepilone, as well as others described herein.
  • ER-related diseases or conditions (for which the agents disclosed herein are therapeutically relevant) include ER-α dysfunction is also associated with cancer (bone cancer, breast cancer, colorectal cancer, endometrial cancer, prostate cancer, ovarian and uterine cancer), leiomyoma (uterine leiomyoma), central nervous system (CNS) defects (alcoholism, migraine), cardiovascular system defects (aortic aneurysm, susceptibility to myocardial infarction, aortic valve sclerosis, cardiovascular disease, coronary artery disease, hypertension), hematological system defects (deep vein thrombosis), immune and inflammation diseases (Graves' Disease, arthritis, multiple sclerosis, cirrhosis), susceptibility to infection (hepatitis B, chronic liver disease), metabolic defects (bone density, cholestasis, hypospadias, obesity, osteoarthritis, osteopenia, osteoporosis), neurological defects (Alzheimer's disease, Parkinson's disease, migraine, vertigo), psychiatric defects (anorexia nervosa, attention deficit hyperactivity disorder (ADHD), dementia, major depressive disorder, psychosis) and reproductive defects (age of menarche, endometriosis, infertility.
  • In some embodiments, compounds disclosed herein are used to treat cancer in a mammal. In some embodiments, the cancer is breast cancer, ovarian cancer, endometrial cancer, prostate cancer, uterine cancer, cervical cancer or lung cancer. In some embodiments, the cancer is breast cancer. In some embodiments, the cancer is a hormone dependent cancer. In some embodiments, the cancer is an estrogen receptor dependent cancer. In some embodiments, the cancer is an estrogen-sensitive cancer. In some embodiments, the cancer is resistant to anti-hormonal treatment. In some embodiments, the cancer is an estrogen-sensitive cancer or an estrogen receptor dependent cancer that is resistant to anti-hormonal treatment. In some embodiments, anti-hormonal treatment includes treatment with at least one agent selected from tamoxifen, fulvestrant, steroidal aromatase inhibitors, and non-steroidal aromatase inhibitors-resistant.
  • In some embodiments, compounds disclosed herein are used to treat hormone receptor positive metastatic breast cancer in a postmenopausal woman with disease progression following anti-estrogen therapy.
  • In some embodiments, compounds disclosed herein are used to treat a hormonal dependent benign or malignant disease of the breast or reproductive tract in a mammal. In some embodiments, the benign or malignant disease is breast cancer.
  • In some embodiments, compounds disclosed herein are used to treat cancer in a mammal, wherein the mammal is chemotherapy-naïve.
  • In some embodiments, compounds disclosed herein are used to treat cancer in a mammal, wherein the mammal is being treated for cancer with at least one anti-cancer agent. In one embodiment, the cancer is a hormone refractory cancer.
  • In some embodiments, compounds disclosed herein are used in the treatment of endometriosis in a mammal.
  • In some embodiments, compounds disclosed herein are used in the treatment of leiomyoma in a mammal. In some embodiments, the leiomyoma is an uterine leiomyoma, esophageal leiomyoma, cutaneous leiomyoma or small bowel leiomyoma. In some embodiments, compounds disclosed herein are used in the treatment of fibroids in a mammal (e.g. uterine fibroids).
  • Compounds
  • Compounds of Formula (I), including pharmaceutically acceptable salts, prodrugs, active metabolites and pharmaceutically acceptable solvates thereof, are estrogen receptor modulators. In specific embodiments, the compounds described herein are estrogen receptor degraders. In specific embodiments, the compounds described herein are estrogen receptor antagonists. In specific embodiments, the compounds described herein are estrogen receptor degraders and estrogen receptor antagonists with minimal or no estrogen receptor agonist activity.
  • In some embodiments, compounds disclosed herein are estrogen receptor degraders and estrogen receptor antagonists that exhibit: minimal or no estrogen receptor agonism; and/or anti-proliferative activity against breast cancer, ovarian cancer, endometrial cancer, cervical cancer cell lines; and/or maximal anti-proliferative efficacy against breast cancer, ovarian cancer, endometrial cancer, cervical cell lines in-vitro; and/or minimal agonism in the human endometrial (Ishikawa) cell line; and/or no agonism in the human endometrial (Ishikawa) cell line; and/or minimal or no agonism in the immature rat uterine assay in-vivo; and/or inverse agonism in the immature rat uterine assay in-vivo; and/or anti-tumor activity in breast cancer, ovarian cancer, endometrial cancer, cervical cancer cell lines in xenograft assays in-vivo or other rodent models of these cancers.
  • In one aspect, provided herein is a compound of Formula (I), or a pharmaceutically acceptable salt, or N-oxide thereof:
  • Figure US20120071535A1-20120322-C00002
      • wherein,
      • Z is —OH or —OR10;
      • R2 is C1-C4alkyl, C1-C4-fluoroalkyl, C1-C4deuteroalkyl, C3-C6cycloalkyl, or —C1-C4alkylene-W;
        • W is hydroxy, halogen, CN, C1-C4alkyl, C1-C4haloalkyl, C1-C4alkoxy, C1-C4haloalkoxy, or C3-C6cycloalkyl;
      • each R3 is independently halogen, C1-C4alkyl, or C1-C4-fluoroalkyl;
      • each R4 is independently halogen, —CN, —OR9, —S(═O)2R10, C1-C4alkyl, C1-C4-fluoroalkyl, or C1-C4heteroalkyl;
      • each R5 is independently halogen, —CN, —OR9, —S(═O)2R10, C1-C4alkyl, C1-C4-fluoroalkyl, or C1-C4heteroalkyl;
      • R6 is H, C1-C4alkyl, or halogen; R7 is H, C1-C4alkyl, or halogen;
      • R9 is H, C1-C6alkyl, C1-C6-fluoroalkyl, or C3-C6cycloalkyl;
      • R10 is C1-C6alkyl;
      • m is 0, 1, or 2; n is 0, 1, 2, 3, or 4; and p is 0, 1, or 2.
  • For any and all of the embodiments, substituents are selected from among from a subset of the listed alternatives. For example, in some embodiments, Z is —OH. In some embodiments, Z is —OR10. In some embodiments, Z is —OH, —OCH3, or —OCH2CH3.
  • In some embodiments, R6 is H, —CH3, F, or Cl. In some embodiments, R6 is H.
  • In some embodiments, R7 is H, —CH3, F, or Cl. In some embodiments, R7 is H.
  • In some embodiments, R9 is H, C1-C6alkyl, or C1-C6-fluoroalkyl. In some embodiments, R9 is H or C1-C6alkyl. In some embodiments, R9 is H.
  • It is understood that R3 may be present on any open position of the indazole ring system. In some embodiments, each R3 is independently halogen, C1-C4alkyl, or C1-C4-fluoroalkyl. In some embodiments, each R3 is independently F, Cl, or —CH3.
  • In some embodiments, each R4 is independently halogen, —CN, —OH, —OR9, —S(═O)2R10, C1-C4alkyl, C1-C4-fluoroalkyl, C1-C4heteroalkyl, C1-C4-fluoroalkoxy, or C1-C4alkoxy. In some embodiments, each R4 is independently halogen, —CN, —OH, —S(═O)2CH3, —S(═O)2CH2CH3, —CH3, —CH2CH3, —CF3, —CH2OH, —OCF3, —OCH3, or —OCH2CH3. In some embodiments, each R4 is independently F, Cl, —CN, —OH, —CH3, —CH2CH3, —CF3, —CH2OH, —OCH3, or —OCH2CH3. In some embodiments, each R4 is independently F or Cl.
  • In some embodiments, each R5 is independently halogen, C1-C4alkyl, or C1-C4-fluoroalkyl. In some embodiments, each R5 is independently F, Cl, or —CH3.
  • In some embodiments, m is 0 or 1. In some embodiments, m is 0. In some embodiments, m is 1.
  • In some embodiments, n is 0, 1, or 2. In some embodiments, n is 0. In some embodiments, n is 1. In some embodiments, n is 2.
  • In some embodiments, p is 0 or 1. In some embodiments, p is 0. In some embodiments, p is 1.
  • In some embodiments, m is 0; and p is 0.
  • In some embodiments, Z is —OH; R6 is H, —CH3, F, or Cl; R7 is H, —CH3, F, or Cl; each R3 is independently halogen, C1-C4alkyl, or C1-C4-fluoroalkyl; each R4 is independently halogen, —CN, —OH, —OR9, —S(═O)2R19, C1-C4alkyl, C1-C4-fluoroalkyl, C1-C4heteroalkyl, C1-C4-fluoroalkoxy, or C1-C4alkoxy; each R5 is independently halogen, C1-C4alkyl, or C1-C4-fluoroalkyl; m is 0 or 1; n is 0, 1, or 2; and p is 0 or 1.
  • In some embodiments, R2 is C1-C4alkyl, C1-C4-fluoroalkyl, C1-C4deuteroalkyl, C3-C6cycloalkyl, or —C1-C4alkylene-W; W is hydroxy, halogen, CN, C1-C4alkoxy, or C3-C6cycloalkyl. In some embodiments, R2 is C1-C4alkyl, C1-C4-fluoroalkyl, or C1-C4deuteroalkyl. In some embodiments, R2 is C1-C4alkyl. In some embodiments, R2 is —CH3, —CH2CH3, —CH2CH2CH3, —CH(CH3)2, —CH2CH2CH2CH3, —CH2CH(CH3)2, —CH2F, —CHF2, —CF3, —CH2CF3, —CD3, —CH2CD3, —CD2CD3, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, —CH2—W, or —CH2CH2—W; W is hydroxy, F, Cl, —CN, —OCH3, —OCH2CH3, —OCH2CH2CH3, —OCH(CH3)2, cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl. In some embodiments, W is hydroxy, F, Cl, —CN, cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl. In some embodiments, R2 is —CH3, —CH2CH3, —CH2CH2CH3, —CH(CH3)2, —CH2CH2CH2CH3, —CH2CH(CH3)2, —CH2F, —CHF2, —CF3, —CH2CF3, —CD3, —CH2CD3, —CD2CD3, —CH2—W, or —CH2CH2—W. In some embodiments, R2 is —CH3, —CH2CH3, —CH2F, —CHF2, —CF3, —CH2CF3, —CD3, —CD2CD3, —CH2CD3, cyclopropyl or cyclobutyl. In some embodiments, R2 is —CH2CH3, —CH2CF3, —CD2CD3, or —CH2CD3. In some embodiments, R2 is —CH2CH3, —CD2CD3, or —CH2CD3. In some embodiments, R2 is —CH2CH3 or cyclobutyl. In some embodiments, R2 is —CH2CH3.
  • In some embodiments, each R3 is independently F, Cl, or —CH3; and m is 0 or 1.
  • In some embodiments, Z is —OH; R6 is H; R7 is H; m is 0; n is 0, 1, or 2; and p is 0.
  • In some embodiments, the compound of Formula (I) has the structure of Formula (II), or a pharmaceutically acceptable salt, or N-oxide thereof:
  • Figure US20120071535A1-20120322-C00003
  • In some embodiments, each R3 is independently F, Cl, or —CH3; each R4 is independently halogen, —CN, —OH, —S(═O)2CH3, —S(═O)2CH2CH3, —CH3, —CH2CH3, —CF3, —CH2OH, —OCF3, —OCH3, or —OCH2CH3; each R5 is independently F, Cl, or —CH3; m is 0 or 1; n is 0, 1, or 2; and p is 0 or 1.
  • In some embodiments, R2 is —CH3, —CH2CH3, —CH2CH2CH3, —CH(CH3)2, —CH2CH2CH2CH3, —CH2CH(CH3)2, —CH2F, —CHF2, —CF3, —CH2CF3, —CD3, —CD2CD3, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, —CH2—W, or —CH2CH2—W; W is hydroxy, F, Cl, —CN, —OCH3, —OCH2CH3, —OCH2CH2CH3, —OCH(CH3)2, cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl.
  • In some embodiments, W is hydroxy, F, Cl, —CN, cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl; each R4 is independently F, Cl, —CN, —OH, —CH3, —CF3, —OCF3, or —OCH3; m is 0 or 1; n is 0, 1, or 2; and p is 0.
  • In some embodiments, R2 is —CH2CH3 or cyclobutyl; each R4 is independently F, Cl, —CN, —OH, —CH3, —CH2CH3, —CF3, —CH2OH, —OCF3, —OCH3, or —OCH2CH3; m is 0 or 1; n is 0, 1, or 2; and p is 0. In some embodiments, R2 is —CH2CH3; each R4 is independently F, Cl, —CN, —OH, —CH3, —CH2CH3, —CF3, —CH2OH, —OCF3, —OCH3, or —OCH2CH3; m is 0; n is 0, 1, or 2; and p is 0.
  • In some embodiments, the compound of Formula (I) or Formula (II) has the following structure, or a pharmaceutically acceptable salt thereof, or N-oxide thereof:
  • Figure US20120071535A1-20120322-C00004
  • In some embodiments,
  • Figure US20120071535A1-20120322-C00005
  • is phenyl, 2-fluorophenyl, 3-fluorophenyl, 4-fluorophenyl, 2,3-difluorophenyl, 2,4-difluorophenyl, 2,5-difluorophenyl, 2,6-difluorophenyl, 2-chlorophenyl, 3-chlorophenyl, 4-chlorophenyl, 2,3-dichlorophenyl, 2,4-dichlorophenyl, 2,5-dichlorophenyl, 2,6-dichlorophenyl, 2-bromophenyl, 3-bromophenyl, 4-bromophenyl, 2-methylphenyl, 3-methylphenyl, 4-methylphenyl, 2-ethylphenyl, 3-ethylphenyl, 4-ethylphenyl, 2-methoxyphenyl, 3-methoxyphenyl, 4-methoxyphenyl, 2-ethoxyphenyl, 3-ethoxyphenyl, 4-ethoxyphenyl, 2-trifluoromethylphenyl, 3-trifluoromethylphenyl, 4-trifluoromethylphenyl, 2-fluoro-4-methoxyphenyl, 2-cyanophenyl, 3-cyanophenyl, 4-cyanophenyl, 2-fluoro-3-chlorophenyl, 2-fluoro-4-chlorophenyl, 2-fluoro-5-chlorophenyl, 2-fluoro-6-chlorophenyl, 2-chloro-3-fluorophenyl, 2-chloro-4-fluorophenyl, 2-chloro-5-fluorophenyl, 2-chloro-6-fluorophenyl, 2-methyl-3-chlorophenyl, 2-methyl-4-chlorophenyl, 2-methyl-5-chlorophenyl, 2-methyl-6-chlorophenyl, 2-methyl-3-fluorophenyl, 2-methyl-4-fluorophenyl, 2-methyl-5-fluorophenyl, 2-methyl-6-fluorophenyl, 3-methyl-4-fluorophenyl, 2-trifluoromethyl-3-chlorophenyl, 2-trifluoromethyl-4-chlorophenyl, 2-trifluoromethyl-5-chlorophenyl, 2-trifluoromethyl-6-chlorophenyl, 2-hydroxyphenyl, 3-hydroxyphenyl, 4-hydroxyphenyl, 2-hydroxymethylphenyl, 3-hydroxymethylphenyl, 4-hydroxymethylphenyl, 2-methylsulfonylphenyl, 3-methylsulfonylphenyl, or 4-methyl sulfonylphenyl.
  • In some embodiments,
  • Figure US20120071535A1-20120322-C00006
  • is phenyl, 2-fluorophenyl, 3-fluorophenyl, 4-fluorophenyl, 2,3-difluorophenyl, 2,4-difluorophenyl, 2,5-difluorophenyl, 2,6-difluorophenyl, 2-chlorophenyl, 3-chlorophenyl, 4-chlorophenyl, 2,3-dichlorophenyl, 2,4-dichlorophenyl, 2,5-dichlorophenyl, 2,6-dichlorophenyl, 2-methylphenyl, 3-methylphenyl, 4-methylphenyl, 2-trifluoromethylphenyl, 3-trifluoromethylphenyl, 4-trifluoromethylphenyl, 2-fluoro-4-methoxyphenyl, 2-fluoro-3-chlorophenyl, 2-fluoro-4-chlorophenyl, 2-fluoro-5-chlorophenyl, 2-fluoro-6-chlorophenyl, 2-chloro-3-fluorophenyl, 2-chloro-4-fluorophenyl, 2-chloro-5-fluorophenyl, 2-chloro-6-fluorophenyl, 2-methyl-3-chlorophenyl, 2-methyl-4-chlorophenyl, 2-methyl-5-chlorophenyl, 2-methyl-6-chlorophenyl, 2-methyl-3-fluorophenyl, 2-methyl-4-fluorophenyl, 2-methyl-5-fluorophenyl, 2-methyl-6-fluorophenyl, 3-methyl-4-fluorophenyl, 2-trifluoromethyl-3-chlorophenyl, 2-trifluoromethyl-4-chlorophenyl, 2-trifluoromethyl-5-chlorophenyl, or 2-trifluoromethyl-6-chlorophenyl.
  • Any combination of the groups described above for the various variables is contemplated herein. Throughout the specification, groups and substituents thereof are chosen by one skilled in the field to provide stable moieties and compounds.
  • In some embodiments, the compound of Formula (I), is a compound presented in Table 1, or a pharmaceutically acceptable salt, or N-oxide thereof:
  • TABLE 1
    LCMS*
    Compound Name Structure [M + H]+
    1 (E)-Ethyl 3-(4-((E)-1-(1H- indazol-5-yl)-2-phenylbut-1- en-1-yl)phenyl)acrylate
    Figure US20120071535A1-20120322-C00007
    423
    2 (E)-3-(4-((E)-1-(1H-Indazol-5- yl)-2-phenylbut-1-en-1- yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00008
    395
    3 (E)-3-(4-((E)-2-(4- Fluorophenyl)-1-(1H-indazol- 5-yl)but-1-en-1- yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00009
    413
    4 (E)-3-(4-((E)-2-(4- Chlorophenyl)-1-(1H-indazol- 5-yl)but-1-en-1- yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00010
    429
    5 (E)-3-(4-((E)-1-(1H-Indazol-5- yl)-2-(3-methoxyphenyl)but-1- en-1-yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00011
    425
    6 (E)-3-(4-((E)-2-(3- (Hydroxymethyl)phenyl)-1- (1H-indazol-5-yl)but-1-en-1- yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00012
    425
    7 (E)-3-(4-((E)-2-(4- (Hydroxymethyl)phenyl)-1- (1H-indazol-5-yl)but-1-en-1- yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00013
    425
    8 (E)-3-(4-((E)-2-(2- (Hydroxymethyl)phenyl)-1- (1H-indazol-5-yl)but-1-en-1- yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00014
    425
    9 (E)-3-(4-((E)-1-(1H-Indazol-5- yl)-2-(o-tolyl)but-1-en-1- yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00015
    409
    10 (E)-3-(4-((E)-1-(1H-Indazol-5- yl)-2-(m-tolyl)but-1-en-1- yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00016
    409
    11 (E)-3-(4-((E)-1-(1H-Indazol-5- yl)-2-(p-tolyl)but-1-en-1- yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00017
    409
    12 (E)-3-(4-((E)-1-(1H-Indazol-5- yl)-2-(2-methoxyphenyl)but-1- en-1-yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00018
    425
    13 (E)-3-(4-((E)-1-(1H-Indazol-5- yl)-2-(4-methoxyphenyl)but-1- en-1-yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00019
    425
    14 ((E)-3-(4-((E)-2-(2- Chlorophenyl)-1-(1H-indazol- 5-yl)but-1-en-1- yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00020
    429
    15 (E)-3-(4-((E)-2-(3- Chlorophenyl)-1-(1H-indazol- 5-yl)but-1-en-1- yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00021
    429
    16 (E)-3-(4-((E)-2-(2- Fluorophenyl)-1-(1H-indazol- 5-yl)but-1-en-1- yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00022
    413
    17 (E)-3-(4-((E)-2-(3- Fluorophenyl)-1-(1H-indazol- 5-yl)but-1-en-1- yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00023
    413
    18 (E)-3-(4-((E)-2-(2- Ethylphenyl)-1-(1H-indazol-5- yl)but-1-en-1- yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00024
    423
    19 (E)-3-(4-((E)-1-(1H-Indazol-5- yl)-2-(2- (trifluoromethyl)phenyl)but-1- en-1-yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00025
    463
    20 (E)-3-(4-((E)-4-Chloro-1-(1H- indazol-5-yl)-2-phenylbut-1- en-1-yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00026
    429
    21 (E)-3-(4-((E)-2-(2- Cyanophenyl)-1-(1H-indazol- 5-yl)but-1-en-1- yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00027
    420
    22 (E)-3-(4-((E)-2-(2,4- Difluorophenyl)-1-(1H- indazol-5-yl)but-1-en-1- yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00028
    431
    23 (E)-3-(4-((E)-2-(2-Chloro-3- fluorophenyl)-1-(1H-indazol- 5-yl)but-1-en-1- yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00029
    447
    24 (E)-3-(4-((E)-2-Cyclopropyl- 1-(1H-indazol-5-yl)-2- phenylvinyl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00030
    407
    25 (E)-3-(4-((E)-2-(4-Fluoro-2- methylphenyl)-1-(1H-indazol- 5-yl)but-1-en-1- yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00031
    427
    26 (E)-3-(4-((E)-2-(2,6- Difluorophenyl)-1-(1H- indazol-5-yl)but-1-en-1- yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00032
    431
    27 (E)-3-(4-((E)-2-(2,6- Dichlorophenyl)-1-(1H- indazol-5-yl)but-1-en-1- yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00033
    463
    28 (E)-3-(4-((E)-4,4,4-Trideutero- 1-(1H-indazol-5-yl)-2- phenylbut-1-en-1- yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00034
    398
    29 (E)-3-(4-((E)-2-(4-Fluoro-3- methylphenyl)-1-(1H-indazol- 5-yl)but-1-en-1- yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00035
    427
    30 (E)-3-(4-((E)-2-(5-Fluoro-2- methylphenyl)-1-(1H-indazol- 5-yl)but-1-en-1- yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00036
    427
    31 (E)-3-(4-((E)-2-(2,3- Difluorophenyl)-1-(1H- indazol-5-yl)but-1-en-1- yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00037
    431
    32 (E)-3-(4-((E)-2-(2,5- Difluorophenyl)-1-(1H- indazol-5-yl)but-1-en-1- yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00038
    431
    33 (E)-3-(4-((E)-2-(2-Chloro-5- fluorophenyl)-1-(1H-indazol- 5-yl)but-1-en-1- yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00039
    447
    34 (E)-3-(4-((E)-2-(2-Chloro-6- methylphenyl)-1-(1H-indazol- 5-yl)but-1-en-1- yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00040
    443
    35 (E)-3-(4-((E)-1-(7-Chloro-1H- indazol-5-yl)-2-phenylbut-1- en-1-yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00041
    429
    36 (E)-3-(4-((E)-1-(4-Methyl-1H- indazol-5-yl)-2-phenylbut-1- en-1-yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00042
    409
    37 (E)-3-(4-((E)-1-(7-Methyl-1H- indazol-5-yl)-2-phenylbut-1- en-1-yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00043
    409
    38 (E)-3-(4-((E)-1-(6-Methyl-1H- indazol-5-yl)-2-phenylbut-1- en-1-yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00044
    409
    39 (E)-3-(4-((E)-1-(3-Methyl-1H- indazol-5-yl)-2-phenylbut-1- en-1-yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00045
    409
    40 (E)-3-(4-((E)-1-(3-Chloro-1H- indazol-5-yl)-2-phenylbut-1- en-1-yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00046
    429
    41 (E)-3-(4-((E)-2-(4-Chloro-2- methylphenyl)-1-(1H-indazol- 5-yl)but-1-en-1- yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00047
    443
    42 (E)-3-(4-((E)-1-(1H-Indazol-5- yl)-2-phenylprop-1-en-1- yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00048
    381
    43 (E)-3-(4-((E)-1-(1H-Indazol-5- yl)-2-phenylpent-1-en-1- yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00049
    409
    44 (E)-3-(4-((E)-2-(3- Cyanophenyl)-1-(1H-indazol- 5-yl)but-1-en-1- yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00050
    420
    45 (E)-3-(4-((E)-2-(4- Cyanophenyl)-1-(1H-indazol- 5-yl)but-1-en-1- yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00051
    420
    46 (E)-3-(4-((E)-4-Hydroxy-1- (1H-indazol-5-yl)-2- phenylbut-1-en-1- yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00052
    411
    47 (E)-3-(4-((E)-1-(1H-Indazol-5- yl)-4-methoxy-2-phenylbut-1- en-1-yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00053
    425
    48 (E)-3-(4-((Z)-1-(1H-Indazol-5- yl)-3-methoxy-2-phenylprop- 1-en-1-yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00054
    411
    49 (E)-3-(4-((E)-1-(4-Fluoro-1H- indazol-5-yl)-2-phenylbut-1- en-1-yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00055
    413
    50 (E)-3-(4-((E)-1-(6-Chloro-1H- indazol-5-yl)-2-phenylbut-1- en-1-yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00056
    429
    51 (E)-3-(4-((E)-1-(1H-Indazol-5- yl)-4-methyl-2-phenylpent-1- en-1-yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00057
    423
    52 (E)-3-(4-((E)-1-(4-Chloro-1H- indazol-5-yl)-2-phenylbut-1- en-1-yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00058
    429
    53 (E)-3-(4-((E)-2-Cyclopentyl-1- (1H-indazol-5-yl)-2- phenylvinyl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00059
    435
    54 (E)-3-(4-((E)-2-Cyclohexyl-1- (1H-indazol-5-yl)-2- phenylvinyl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00060
    449
    55 (E)-3-(4-((E)-1-(1H-Indazol-5- yl)-3-methyl-2-phenylbut-1- en-1-yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00061
    409
    56 (E)-3-(4-((E)-3-Cyclopropyl- 1-(1H-indazol-5-yl)-2- phenylprop-1-en-1- yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00062
    421
    57 (E)-3-(4-((E)-2-(2- Chlorophenyl)-2-cyclopropyl- 1-(1H-indazol-5- yl)vinyl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00063
    441
    58 (E)-3-(4-((E)-1-(6-Fluoro-1H- indazol-5-yl)-2-phenylbut-1- en-1-yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00064
    413
    59 (E)-3-(4-((E)-1-(1H-Indazol-5- yl)-2-phenylhex-1-en-1- yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00065
    423
    60 (E)-3-(4-((E)-3-Cyclopentyl-1- (1H-indazol-5-yl)-2- phenylprop-1-en-1- yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00066
    449
    61 (E)-3-(4-((E)-2-(2-Chloro-4- fluorophenyl)-1-(4-fluoro-1H- indazol-5-yl)but-1-en-1- yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00067
    465
    62 (E)-3-(4-((E)-1-(7-Fluoro-1H- indazol-5-yl)-2-phenylbut-1- en-1-yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00068
    413
    63 (E)-3-(4-((E)-2-(2-Chloro-4- fluorophenyl)-1-(1H-indazol- 5-yl)-4-methylpent-1-en-1- yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00069
    475
    64 (E)-3-(4-((Z)-3,3-Difluoro-1- (1H-indazol-5-yl)-2- phenylprop-1-en-1- yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00070
    417
    65 (E)-3-(4-((E)-2-(2-Chloro-4- fluorophenyl)-1-(7-fluoro-1H- indazol-5-yl)but-1-en-1- yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00071
    465
    66 (E)-3-(4-((E)-4-Fluoro-1-(1H- indazol-5-yl)-2-phenylbut-1- en-1-yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00072
    413
    67 (E)-3-(4-((E)-4-Chloro-2-(2- chloro-4-fluorophenyl)-1-(1H- indazol-5-yl)but-1-en-1- yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00073
    481
    68 (E)-3-(4-((Z)-3,3,3-Trifluoro- 1-(1H-indazol-5-yl)-2- phenylprop-1-en-1- yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00074
    not observed
    69 (E)-3-(4-((E)-1-(4-Fluoro-1H- indazol-5-yl)-2-(4-fluoro-2- methylphenyl)but-1-en-1- yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00075
    445
    70 (E)-3-(4-((E)-2-(4-Chloro-2- methylphenyl)-1-(4-fluoro-1H- indazol-5-yl)but-1-en-1- yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00076
    461
    71 (E)-3-(4-((E)-2-Cyclopropyl- 1-(4-fluoro-1H-indazol-5-yl)- 2-(4-fluoro-2- methylphenyl)vinyl)phenyl) acrylic acid
    Figure US20120071535A1-20120322-C00077
    457
    72 (E)-3-(4-((E)-2-(4-Chloro-2- methylphenyl)-2- cyclopropyl- 1-(4-fluoro-1H-indazol-5- yl)vinyl)phenyl) acrylic acid
    Figure US20120071535A1-20120322-C00078
    473
    73 (E)-3-(4-((E)-1-(4-Chloro-1H- indazol-5-yl)-2-(2-chloro-4- fluorophenyl)but-1-en-1- yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00079
    481
    74 (E)-3-(4-((Z)-2-(2-Chloro-4- fluorophenyl)-3,3-difluoro-1- (1H-indazol-5-yl)prop-1-en-1- yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00080
    469
    75 (E)-3-(4-((E)-2-Cyclopropyl- 1-(4-fluoro-1H-indazol-5-yl)- 2-phenylvinyl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00081
    425
    76 (E)-3-(4-((E)-4-Chloro-1-(4- fluoro-1H-indazol-5-yl)-2- phenylbut-1-en-1- yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00082
    447
    77 (E)-3-(4-((E)-4-Chloro-2-(2- chloro-4-fluorophenyl)-1-(4- fluoro-1H-indazol-5-yl)but-1- en-1-yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00083
    499
    78 (E)-3-(4-((E)-4-Fluoro-2-(4- fluorophenyl)-1-(1H-indazol- 5-yl)but-1-en-1- yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00084
    431
    79 (E)-3-(4-((E)-4-Fluoro-1-(4- fluoro-1H-indazol-5-yl)-2- phenylbut-1-en-1- yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00085
    431
    80 (E)-3-(4-((E)-1-(1H-Indazol-5- yl)-5-methoxy-2-phenylpent- 1-en-1-yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00086
    439
    81 (E)-3-(4-((E)-1-(1H-Indazol-5- yl)-6-methoxy-2-phenylhex-1- en-1-yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00087
    453
    82 (E)-3-(4-((E)-2-(2-Chloro-4- fluorophenyl)-1-(1H-indazol- 5-yl)-3-methylbut-1-en-1- yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00088
    461
    83 (E)-3-(4-((E)-1-(1H-Indazol-5- yl)-2-(3- (trifluoromethoxy)phenyl)but- 1-en-1-yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00089
    479
    84 (E)-3-(4-((E)-2-Cyclobutyl-1- (1H-indazol-5-yl)-2- phenylvinyl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00090
    421
    85 (E)-3-(4-((E)-2-(2-Chloro-4- fluorophenyl)-2-cyclobutyl-1- (1H-indazol-5- ypvinyl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00091
    473
    86 (E)-3-(4-((E)-1-(3-Fluoro-1H- indazol-5-yl)-2-phenylbut-1- en-1-yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00092
    413
    87 (E)-3-(4-((E)-2-Cyclobutyl-1- (3-fluoro-1H-indazol-5-yl)-2- phenylvinyl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00093
    439
    88 (E)-3-(4-((E)-2-(2-Chloro-4- fluorophenyl)-1-(3-fluoro-1H- indazol-5-yl)but-1-en-1- yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00094
    465
    89 (E)-3-(4-((E)-2-(2-Chloro-4- fluorophenyl)-2-cyclobutyl-1- (3-fluoro-1H-indazol-5- yl)vinyl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00095
    491
    90 (E)-Ethyl 3-(4-((E)-2-(4- fluorophenyl)-1-(1H-indazol- 5-yl)but-1-en-1- yl)phenyl)acrylate
    Figure US20120071535A1-20120322-C00096
    441
    91 (E)-Ethyl 3-(4-((E)-1-(1H- Indazol-5-yl)-2-(2- methoxyphenyl)but-1-en-1- yl)phenyl)acrylate
    Figure US20120071535A1-20120322-C00097
    453
    92 (E)-Ethyl 3-(4-((E)-1-(1H- Indazol-5-yl)-2-(4- methoxyphenyl)but-1-en-1- yl)phenyl)acrylate
    Figure US20120071535A1-20120322-C00098
    453
    93 (E)-3-(4-((E)-2-(3- Hydroxyphenyl)-1-(1H- indazol-5-yl)but-1-en-1- yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00099
    411
    94 (E)-3-(4-((E)-2-(2- Hydroxyphenyl)-1-(1H- indazol-5-yl)but-1-en-1- yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00100
    411
    95 (E)-3-(4-((E)-2-(4- Hydroxyphenyl)-1-(1H- indazol-5-yl)but-1-en-1- yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00101
    411
    96 (E)-3-(4-((E)-2-(3- Butoxyphenyl)-1-(1H-indazol- 5-yl)but-1-en-1- yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00102
    467
    97 (E)-3-(4-((E)-2-(4- Butoxyphenyl)-1-(1H-indazol- 5-yl)but-1-en-1- yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00103
    467
    98 (E)-3-(4-((E)-1-(1H-Indazol-5- yl)-2-(2- (methylsulfonyl)phenyl)but-1- en-1-yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00104
    473
    99 (E)-3-(4-((E)-1-(1H-Indazol-5- yl)-2-phenylbut-1-en-1- yl)phenyl)-2-methylacrylic acid
    Figure US20120071535A1-20120322-C00105
    409
    100 (E)-3-(4-((Z)-1-(1H-Indazol-5- yl)-2-phenylbut-1-en-1-yl)-3- methylphenyl)acrylic acid
    Figure US20120071535A1-20120322-C00106
    409
    101 (E)-3-(4-((E)-1-(1H-Indazol-5- yl)-2-phenylbut-1-en-1-yl)-2- methylphenyl)acrylic acid
    Figure US20120071535A1-20120322-C00107
    409
    102 (E)-3-(4-((Z)-1-(1H-Indazol-5- yl)-2-phenylbut-1-en-1-yl)-2- chlorophenyl)acrylic acid
    Figure US20120071535A1-20120322-C00108
    429
    103 (Z)-3-(4-((E)-1-(1H-Indazol-5- yl)-2-phenylbut-1-en-1- yl)phenyl)-2-fluoroacrylic acid
    Figure US20120071535A1-20120322-C00109
    413
    104 (Z)-3-(4-((E)-1-(1H-Indazol-5- yl)-2-phenylbut-1-en-1- yl)phenyl)-2-chloroacrylic acid
    Figure US20120071535A1-20120322-C00110
    429
    105 (E)-3-(4-((Z)-1-(1H-Indazol-5- yl)-2-phenylbut-1-en-1-yl)-3- fluorophenyl)acrylic acid
    Figure US20120071535A1-20120322-C00111
    413
    106 (E)-3-(4-((Z)-1-(1H-Indazol-5- yl)-2-phenylbut-1-en-1-yl)-2- fluorophenyl)acrylic acid
    Figure US20120071535A1-20120322-C00112
    413
    107 (E)-3-(4-((Z)-1-(1H-Indazol-5- yl)-2-phenylbut-1-en-1-yl)-2- (trifluoromethyl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00113
    463
    108 (E)-3-(4-((Z)-1-(1H-Indazol-5- yl)-2-phenylbut-1-en-1-yl)-3- methoxyphenyl)acrylic acid
    Figure US20120071535A1-20120322-C00114
    425
    109 (E)-3-(4-((Z)-1-(1H-Indazol-5- yl)-2-phenylbut-1-en-1-yl)-2- methoxyphenyl)acrylic acid
    Figure US20120071535A1-20120322-C00115
    425
    110 (E)-Ethyl 3-(4-((E)-2-(2- chloro-4-fluorophenyl)-1-(1H- indazol-5-yl)but-1-en-1- yl)phenyl)acrylate hydrochloride
    Figure US20120071535A1-20120322-C00116
    475
    111 (E)-3-(4-((E)-2-(2-Chloro-4- fluorophenyl)-1-(1H-indazol- 5-yl)but-1-en-1- yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00117
    447
    112 (E)-Ethyl 3-(4-((E)-2-(2,4- dichlorophenyl)-1-(1H- indazol-5-yl)but-1-en-1- yl)phenyl)acrylate
    Figure US20120071535A1-20120322-C00118
    491
    113 (E)-3-(4-((E)-2-(2,4- Dichlorophenyl)-1-(1H- indazol-5-yl)but-1-en-1- yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00119
    463
    114 (E)-3-(4-((E)-2-(4-Chloro-2- (trifluoromethyl)phenyl)-1- (1H-indazol-5-yl)but-1-en-1- yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00120
    497
    115 (E)-3-(4-((E)-2-(2-Chloro-4- fluorophenyl)-2-cyclopropyl- 1-(1H-indazol-5- yl)vinyl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00121
    459
    116 (E)-3-(4-((E)-2-(4-Fluoro-2- (trifluoromethyl)phenyl)-1- (1H-indazol-5-yl)but-1-en-1- yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00122
    481
    117 (E)-3-(4-(1-(4-Fluoro-1H- indazol-5-yl)-2-(4-fluoro-2- (trifluoromethyl) phenyl)butyl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00123
    499
    118 (E)-3-(4-((E)-2-(2,4- Dichlorophenyl)-1-(4-fluoro- 1H-indazol-5-yl)but-1-en-1- yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00124
    481
    119 (E)-3-(4-((E)-2-(4-Chloro-2- (trifluoromethyl)phenyl)-1-(4- fluoro-1H-indazol-5-yl)but-1- en-1-yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00125
    515
    120 (E)-3-(4-((E)-2-(2-Chloro-4- fluorophenyl)-4-fluoro-1-(1H- indazol-5-yl)but-1-en-1- yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00126
    465
    121 (E)-3-(4-((E)-2-(2-Chloro-4- methoxyphenyl)-1-(1H- indazol-5-yl)but-1-en-1- yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00127
    459
    122 (E)-3-(4-((E)-2-(2,4- Dichlorophenyl)-4-fluoro-1- (4-fluoro-1H-indazol-5-yl)but- 1-en-1-yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00128
    499
    123 (E)-3-(4-((E)-2-Cyclopropyl- 2-(2,4-dichlorophenyl)-1-(4- fluoro-1H-indazol-5- yl)vinyl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00129
    493
    124 (E)-3-(4-((E)-2-(2-Chloro-4- fluorophenyl)-2-cyclopropyl- 1-(4-fluoro-1H-indazol-5- yl)vinyl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00130
    477
    125 (E)-3-(4-((E)-2-Cyclopropyl- 2-(2,4-dichlorophenyl)-1-(1H- indazol-5- yl)vinyl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00131
    475
    126 (E)-3-(4-((E)-2-(4-Chloro-2- methylphenyl)-2-cyclopropyl- 1-(1H-indazol-5- yl)vinyl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00132
    455
    127 (E)-3-(4-((E)-1-(1H-Indazol-5- yl)-2-(2-methyl-5- (methylsulfonyl)phenyl)but-1- en-1-yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00133
    487
    128 (E)-3-(4-((E)-1-(1H-Indazol-5- yl)-2-(4-methoxy-2- methylphenyl)but-1-en-1- yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00134
    439
    129 (E)-3-(4-((E)-2-(2-Fluoro-4- methoxyphenyl)-1-(1H- indazol-5-yl)but-1-en-1- yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00135
    443
    130 (E)-3-(4-((E)-2-(2-Chloro-5- methoxyphenyl)-1-(1H- indazol-5-yl)but-1-en-1- yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00136
    459
    131 (E)-3-(4-((E)-2-(2-Fluoro-4- (methylsulfonyl)phenyl)-1- (1H-indazol-5-yl)but-1-en-1- yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00137
    491
    132 (E)-3-(4-((E)-2-(2,4- Dichlorophenyl)-3,3,4,4,4- pentadeutero-1-(1H-indazol-5- yl)but-1-en-1- yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00138
    468
    133 (E)-3-(4-((E)-1-(1H-Indazol-5- yl)-2-(3- (methylsulfonyl)phenyl)but-1- en-1-yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00139
    473
    134 (E)-3-(4-((E)-2-(2,4- Dichlorophenyl)-1-(7-fluoro- 1H-indazol-5-yl)but-1-en-1- yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00140
    481
    135 (E)-3-(4-((E)-2-(2-Chloro-3- methoxyphenyl)-1-(1H- indazol-5-yl)but-1-en-1- yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00141
    459
    136 (E)-3-(4-((E)-2-(2-Chloro-4- fluorophenyl)-3,3,4,4,4- pentadeutero-1-(1H-indazol-5- yl)but-1-en-1- yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00142
    452
    137 (E)-3-(4-((E)-2-(4-Chloro-2- cyanophenyl)-1-(1H-indazol- 5-yl)but-1-en-1- yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00143
    454
    138 (E)-3-(4-((E)-2-(2-Cyano-4- fluorophenyl)-1-(1H-indazol- 5-yl)but-1-en-1- yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00144
    438
    139 (E)-3-(4-((E)-2-(2-Cyano-4- (trifluoromethyl)phenyl)-1- (1H-indazol-5-yl)but-1-en-1- yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00145
    488
    140 (E)-3-(4-((E)-2-(2-Chloro-4- cyanophenyl)-1-(1H-indazol- 5-yl)but-1-en-1- yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00146
    454
    141 (E)-3-(4-((E)-2-(3-Cyano-2- methylphenyl)-1-(1H-indazol- 5-yl)but-1-en-1- yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00147
    434
    142 (E)-3-(4-((E)-2-(4-Cyano-2- methylphenyl)-1-(1H-indazol- 5-yl)but-1-en-1- yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00148
    434
    143 (E)-3-(4-((E)-2-(5-Cyano-2- methylphenyl)-1-(1H-indazol- 5-yl)but-1-en-1- yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00149
    434
    144 (E)-3-(4-((E)-2-(2-Cyano-4- methoxyphenyl)-1-(1H- indazol-5-yl)but-1-en-1- yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00150
    450
    145 (E)-3-(4-((E)-2-(2- Chlorophenyl)-1-(1-methyl- 1H-indazol-5-yl)but-1-en-1- yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00151
    443
    146 (E)-3-(4-((E)-2-Cyclobutyl-1- (1-methyl-1H-indazol-5-yl)-2- phenylvinyl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00152
    435
    147 (E)-3-(4-((E)-2-(2-Chloro-4- fluorophenyl)-1-(1-methyl-1H- indazol-5-yl)but-1-en-1- yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00153
    461
    148 (E)-3-(4-((E)-2-(2-Chloro-4- fluorophenyl)-1-(1- (difluoromethyl)-1H-indazol- 5-yl)but-1-en-1- yl)phenyl)acrylic acid
    Figure US20120071535A1-20120322-C00154
    497
    *mass spectrometric data
  • Synthesis of Compounds
  • Compounds of Formula (I), described herein, are synthesized using standard synthetic techniques or using methods known in the art in combination with methods described herein. In addition, solvents, temperatures and other reaction conditions presented herein may vary.
  • The starting material used for the synthesis of the compounds of Formula (I) are either synthesized or obtained from commercial sources, such as, but not limited to, Sigma-Aldrich, Fluka, Acros Organics, Alfa Aesar, and the like. The compounds described herein, and other related compounds having different substituents are synthesized using techniques and materials described herein or otherwise known, including those found in March, ADVANCED ORGANIC CHEMISTRY 4th Ed., (Wiley 1992); Carey and Sundberg, ADVANCED ORGANIC CHEMISTRY 4th Ed., Vols. A and B (Plenum 2000, 2001), and Green and Wuts, PROTECTIVE GROUPS IN ORGANIC SYNTHESIS 3rd Ed., (Wiley 1999). General methods for the preparation of compounds is optionally modified by the use of appropriate reagents and conditions for the introduction of the various moieties found in the formulae as provided herein.
  • In some embodiments, exemplary compounds of Formula (I) are prepared as outlined in the following Schemes.
  • Figure US20120071535A1-20120322-C00155
  • Protection of the acidic proton bearing nitrogen of compounds of structure 1 with a protecting group provides compounds of Structure 2. In some embodiments, the protecting group is tetrahydro-2H-pyran (THP). In some embodiments, the conditions for nitrogen protection require 3,4-dihydro-2H-pyran (DHP), an organic acid and a suitable solvent. In some embodiments, the organic acid is pyridinium p-toluenesulfonate (PPTS) or p-toluenesulfonic acid (p-TSA or p-TsOH) and the suitable solvent is dichloromethane. In some embodiments, the reaction is performed at room temperature. Other conditions to protect the nitrogen of the starting material are known. A detailed description of techniques applicable to the creation of protecting groups and their removal are described in Greene and Wuts, Protective Groups in Organic Synthesis, 3rd Ed., John Wiley & Sons, New York, N.Y., 1999, and Kocienski, Protective Groups, Thieme Verlag, New York, N.Y., 1994, which are incorporated herein by reference for such disclosure.
  • Compounds of structure 2, where X is a halogen or other suitable leaving group, are reacted with a protected acetylene (e.g., trimethylsilylacetylene) under Sonogashira reaction conditions to provide compounds of Structure 3. In some embodiments, the Sonogashira coupling reaction conditions include the use of a palladium catalyst and a copper halide salt. In some embodiments, the Sonogashira reaction conditions in the use of Pd(Ph3P)2Cl2, CuI, and triethylamine. In one embodiment, the reaction is performed at about 80° C. Other suitable reaction conditions are described in Rafael Chinchilla and Carmen Nájera (2007). Chem. Rev. 107 (3): 874-922.
  • The silyl protecting group of compounds of structure 3 is removed under suitable reaction conditions to provide compounds of structure 4. In some embodiments, the silyl protecting group is removed with potassium carbonate (K2CO3) in methanol. In other embodiments, the silyl protecting group is removed with tetrabutylammoniumfluoride (TBAF) in tetrahydrofuran.
  • In some embodiments, acetylenes of Structure 4 are reacted with R2—X under basic condition to prepare compounds of Structure 5. In these instances, R2 is C1-C6alkyl or C1-C6-fluoroalkyl or C1-C4alkoxy or C1-C4-fluoroalkoxy or C3-C6cycloalkyl, or the like, and X is a suitable leaving group. In some embodiments, R2 moieties are installed by other suitable conditions.
  • In some embodiments, compounds of structure 2 are coupled with an alkynyl-trimethylsilane or a terminal-alkyne under Sonogashira reaction conditions to provide compounds of structure 5. In some embodiments, the coupling of an alkynyl-trimethylsilane with compounds of structure 2 includes the use of a base (e.g. cesium carbonate), a palladium catalyst (e.g. Pd(OAc)2, dppf) and a copper halide salt (e.g. CuI) in a suitable solvent (e.g. dimethylacetamide), at elevated temperatures (e.g. about 80-90° C.). In some embodiments, the coupling of a terminal-alkyne with compounds of structure 2 includes the use of Pd(PPh3)2Cl2), CuI, and triethylamine, with the reaction performed with at elevated temperatures (e.g. about 80-120° C.).
  • Figure US20120071535A1-20120322-C00156
  • Compounds of Structures 5, 6 and 7 are then coupled together under suitable reaction conditions to afford compounds of Structure 8. In some embodiments, the suitable reaction conditions include the use of organometallic reagent(s). In some embodiments, the suitable reaction conditions include the use of a palladium catalyst. In some embodiments, the suitable reaction conditions include the use of Pd(PhCN)2Cl2, K2CO3 in dimethylformamide/water. Other suitable reaction conditions include those described in Chengxiang Zhou and Richard C. Larock, Journal of Organic Chemistry, 2005, 70, 3765-3777; Chengxiang Zhou, Daniel E. Emrich, and Richard C. Larock Organic Letters 2003, 1579-1582; Tsutomu Konno, Ken-ichi Taku, Takashi Ishihara, Journal of Fluorine Chemistry 127 (2006) 966-972.
  • The protecting group of compounds of structure 8 is then removed under suitable reaction conditions to provide compounds of structure 9. In some embodiments, the suitable reaction conditions include the use of an acid. In some embodiments, the suitable reaction conditions include the use of hydrochloric acid, ethanol, with the reaction performed at about 70° C.
  • Hydrolysis of the ester group of compounds of structure 9 provides carboxylic acid compounds of structure 10. In some embodiments, the hydrolysis reaction includes the use of lithium hydroxide in a mixture of tetrahydrofuran and ethanol. Other hydrolysis reaction conditions are known.
  • In some embodiments, compounds disclosed herein are prepared as outlined in Scheme 3.
  • Figure US20120071535A1-20120322-C00157
  • In some embodiments, compounds of structure 5 are reacted with phenyl halides of structure 6 and boronic acids of structure 11 under suitable reaction conditions to provide compounds of structure 12. In some embodiments, the suitable reaction conditions include the use of organometallic reagent(s). In some embodiments, the suitable organometallic reagent is a palladium catalyst. The aldehyde of compounds of structure 12 is then transformed to an alkene under suitable reaction conditions to provide compounds of structure 8. Suitable reaction conditions include a Horner-Wadsworth-Emmons olefination reaction or a Wittig olefination reaction conditions.
  • Alternatively, compounds of structure 5 are reacted with a borylating agent in the presence of a suitable catalyst to provide compounds of structure 13. In some embodiments, the suitable catalyst is an organometallic reagent such as a platinum catalyst. In some embodiments, the amount of catalyst impacts the rate of the reaction, but generally, not the yield or purity. In some embodiments, the solvent has a small impact on the rate of the reaction, but generally, not the yield or purity. In some embodiments, the temperature has a significant impact on the rate of the reaction, but generally, not the yield or purity. A Suzuki cross-coupling is then performed with compounds of structure 13 and phenyl halides of structure 14 to provide compounds of structure 15. In some embodiments, 2 or 3 equivalents of base (e.g. Cs2CO3) is used in the Suzuki cross-coupling. In some embodiments, 1.3 equivalents of base (e.g. Cs2CO3) is used in the Suzuki cross-coupling. In some embodiments, the solvent has a significant impact on the rate and regioselectivity of this reaction. In some embodiments, dioxane, DME, or 2-MeTHF is used. In some embodiments, water content has a significant impact on the rate and regioselectivity of this the Suzuki cross-coupling. A subsequent Suzuki cross-coupling is then performed between compounds of structure 15 and phenyl halides of structure 16 to provide compounds of structure 12.
  • A detailed description of techniques applicable to the creation of protecting groups and their removal are described in Greene and Wuts, Protective Groups in Organic Synthesis, 3rd Ed., John Wiley & Sons, New York, N.Y., 1999, and Kocienski, Protective Groups, Thieme Verlag, New York, N.Y., 1994, which are incorporated herein by reference for such disclosure.
  • Further Forms of Compounds
  • In instances where the compound of Formula (I) possesses one or more stereocenters, each stereocenter exists independently in either the R or S configuration. The compounds presented herein include all diastereomeric, enantiomeric, atropisomers, and epimeric forms as well as the appropriate mixtures thereof. The compounds and methods provided herein include all cis, trans, syn, anti, entgegen (E), and zusammen (Z) isomers as well as the appropriate mixtures thereof.
  • Stereoisomers are obtained, if desired, by methods such as, stereoselective synthesis and/or the separation of stereoisomers by chiral chromatographic columns and/or use of optically active resolving agents. Jean Jacques, Andre Collet, Samuel H. Wilen, “Enantiomers, Racemates and Resolutions”, John Wiley And Sons, Inc., 1981.
  • In certain embodiments, the compounds presented herein are present as atropisomers. Atropisomers refer to stereoisomers resulting from hindered rotation about single bonds where the steric strain barrier to rotation allows for the isolation of conformers. Atropisomers display axial chirality. Separation of atropisomers is possible. In some embodiments, separation of atropisomers is possible by chiral resolution methods such as selective crystallization. Atropisomers are optionally characterized by NMR or other suitable characterization means.
  • The methods and compositions described herein include the use of amorphous forms as well as crystalline forms (also known as polymorphs). In one aspect, compounds described herein are in the form of pharmaceutically acceptable salts. As well, active metabolites of these compounds having the same type of activity are included in the scope of the present disclosure. In addition, the compounds described herein can exist in unsolvated as well as solvated forms with pharmaceutically acceptable solvents such as water, ethanol, and the like. The solvated forms of the compounds presented herein are also considered to be disclosed herein.
  • In some embodiments, compounds described herein are prepared as prodrugs. A “prodrug” refers to an agent that is converted into the parent drug in vivo. Prodrugs are often useful because, in some situations, they are easier to administer than the parent drug or they are bioavailable by oral administration or they have improved solubility in pharmaceutical compositions over the parent drug. In some embodiments, the design of a prodrug increases the effective water solubility. An example, without limitation, of a prodrug is a compound described herein, which is administered as an ester (the “prodrug”) but then is metabolically hydrolyzed to provide the active entity. In certain embodiments, upon in vivo administration, a prodrug is chemically converted to the biologically, pharmaceutically or therapeutically active form of the compound.
  • In some embodiments, sites on the aromatic ring portion of compounds of Formula (I) are susceptible to various metabolic reactions. Incorporation of appropriate substituents on the aromatic ring structures will reduce, minimize or eliminate this metabolic pathway. In specific embodiments, the appropriate substituent to decrease or eliminate the susceptibility of the aromatic ring to metabolic reactions is, by way of example only, a halogen, deuterium or an alkyl group.
  • In another embodiment, the compounds described herein are labeled isotopically (e.g. with a radioisotope) or by another other means, including, but not limited to, the use of chromophores or fluorescent moieties, bioluminescent labels, or chemiluminescent labels.
  • In additional or further embodiments, the compounds described herein are metabolized upon administration to an organism in need to produce a metabolite that is then used to produce a desired effect, including a desired therapeutic effect.
  • “Pharmaceutically acceptable,” as used herein, refers to a material, such as a carrier or diluent, which does not abrogate the biological activity or properties of the compound, and is relatively nontoxic, i.e., the material is administered to an individual without causing undesirable biological effects or interacting in a deleterious manner with any of the components of the composition in which it is contained.
  • The term “pharmaceutically acceptable salt” refers to a formulation of a compound that does not cause significant irritation to an organism to which it is administered and does not abrogate the biological activity and properties of the compound. In some embodiments, pharmaceutically acceptable salts are obtained by reacting a compound of Formula (I) with an acid. Pharmaceutically acceptable salts are also obtained by reacting a compound of Formula (I) with a base to form a salt.
  • Compounds described herein are optionally formed as, and/or used as, pharmaceutically acceptable salts. The type of pharmaceutical acceptable salts, include, but are not limited to: (1) acid addition salts, formed by reacting the free base form of the compound with a pharmaceutically acceptable: inorganic acid, such as, for example, hydrochloric acid, hydrobromic acid, sulfuric acid, phosphoric acid, metaphosphoric acid, and the like; or with an organic acid, such as, for example, acetic acid, propionic acid, hexanoic acid, cyclopentanepropionic acid, glycolic acid, pyruvic acid, lactic acid, malonic acid, succinic acid, malic acid, maleic acid, fumaric acid, trifluoroacetic acid, tartaric acid, citric acid, benzoic acid, 3-(4-hydroxybenzoyl)benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, 1,2-ethanedisulfonic acid, 2-hydroxyethanesulfonic acid, benzenesulfonic acid, toluenesulfonic acid, 2-naphthalenesulfonic acid, 4-methylbicyclo-[2.2.2]oct-2-ene-1-carboxylic acid, glucoheptonic acid, 4,4′-methylenebis-(3-hydroxy-2-ene-1-carboxylic acid), 3-phenylpropionic acid, trimethylacetic acid, tertiary butylacetic acid, lauryl sulfuric acid, gluconic acid, glutamic acid, hydroxynaphthoic acid, salicylic acid, stearic acid, muconic acid, butyric acid, phenylacetic acid, phenylbutyric acid, valproic acid, and the like; (2) salts formed when an acidic proton present in the parent compound is replaced by a metal ion, e.g., an alkali metal ion (e.g. lithium, sodium, potassium), an alkaline earth ion (e.g. magnesium, or calcium), or an aluminum ion. In some cases, compounds described herein coordinate with an organic base, such as, but not limited to, ethanolamine, diethanolamine, triethanolamine, tromethamine, N-methylglucamine, dicyclohexylamine, tris(hydroxymethyl)methylamine. In other cases, compounds described herein form salts with amino acids such as, but not limited to, arginine, lysine, and the like. Acceptable inorganic bases used to form salts with compounds that include an acidic proton, include, but are not limited to, aluminum hydroxide, calcium hydroxide, potassium hydroxide, sodium carbonate, sodium hydroxide, and the like. In some embodiments, the compounds provided herein are prepared as lysine salts, sodium salts and other suitable amino acid salts. In some embodiments, the compounds provided herein are prepared as a sodium salt. In some embodiments, the compounds provided herein are prepared as an N-methylglucamine salt. In some embodiments, the compounds provided herein are prepared as a hydrochloride salt.
  • It should be understood that a reference to a pharmaceutically acceptable salt includes the solvent addition forms. In some embodiments, solvates contain either stoichiometric or non-stoichiometric amounts of a solvent, and are formed during the process of crystallization with pharmaceutically acceptable solvents such as water, ethanol, and the like. Hydrates are formed when the solvent is water, or alcoholates are formed when the solvent is alcohol. Solvates of compounds described herein are conveniently prepared or formed during the processes described herein. In addition, the compounds provided herein optionally exist in unsolvated as well as solvated forms.
  • The methods and formulations described herein include the use of N-oxides (if appropriate), crystalline forms (also known as polymorphs), or pharmaceutically acceptable salts of compounds having the structure of Formula (I), as well as active metabolites of these compounds having the same type of activity.
  • Certain Terminology
  • Unless otherwise stated, the following terms used in this application, including the specification and claims, have the definitions given below. It must be noted that, as used in the specification and the appended claims, the singular forms “a,” “an” and “the” include plural referents unless the context clearly dictates otherwise. Unless otherwise indicated, conventional methods of mass spectroscopy, NMR, HPLC, protein chemistry, biochemistry, recombinant DNA techniques and pharmacology are employed. In this application, the use of “or” or “and” means “and/or” unless stated otherwise. Furthermore, use of the term “including” as well as other forms, such as “include”, “includes,” and “included,” is not limiting. The section headings used herein are for organizational purposes only and are not to be construed as limiting the subject matter described.
  • An “alkyl” group refers to an aliphatic hydrocarbon group. The alkyl group is saturated or unsaturated. The alkyl moiety, whether saturated or unsaturated, is branched or straight chain. In one aspect the alkyl is selected from the group consisting of methyl, ethyl, propyl, iso-propyl, n-butyl, iso-butyl, sec-butyl, and t-butyl. Typical alkyl groups include, but are in no way limited to, methyl, ethyl, propyl, isopropyl, butyl, isobutyl, sec-butyl, tertiary butyl, pentyl, neopentyl, hexyl, allyl, vinyl, acetylene, but-2-enyl, but-3-enyl, and the like.
  • “Deuteroalkyl” refers to an alkyl group where 1 or more hydrogen atoms of an alkyl are replaced with deuterium.
  • An “alkoxy” group refers to a (alkyl)O— group, where alkyl is as defined herein.
  • The term “cycloalkyl” refers to cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl or cyclohexenyl.
  • The term “halo” or, alternatively, “halogen” or “halide” means fluoro, chloro, bromo or iodo.
  • The term “fluoroalkyl” refers to an alkyl in which one or more hydrogen atoms are replaced by a fluorine atom.
  • The term “heteroalkyl” refers to an alkyl group in which one or more skeletal atoms of the alkyl are selected from an atom other than carbon, e.g., oxygen, nitrogen (e.g. —NH—, —N(alkyl)-, sulfur, or combinations thereof. In one aspect, a heteroalkyl is a C1-C6heteroalkyl.
  • The term “acceptable” with respect to a formulation, composition or ingredient, as used herein, means having no persistent detrimental effect on the general health of the subject being treated.
  • The term “modulate” as used herein, means to interact with a target either directly or indirectly so as to alter the activity of the target, including, by way of example only, to enhance the activity of the target, to inhibit the activity of the target, to limit the activity of the target, or to extend the activity of the target.
  • The term “modulator” as used herein, refers to a molecule that interacts with a target either directly or indirectly. The interactions include, but are not limited to, the interactions of an agonist, partial agonist, an inverse agonist, antagonist, degrader, or combinations thereof. In some embodiments, a modulator is an antagonist. In some embodiments, a modulator is a degrader.
  • “Selective estrogen receptor modulator” or “SERM” as used herein, refers to a molecule that differentially modulates the activity of estrogen receptors in different tissues. For example, in some embodiments, a SERM displays ER antagonist activity in some tissues and ER agonist activity in other tissues. In some embodiments, a SERM displays ER antagonist activity in some tissues and minimal or no ER agonist activity in other tissues. In some embodiments, a SERM displays ER antagonist activity in breast tissues, ovarian tissues, endometrial tissues, and/or cervical tissues but minimal or no ER agonist activity in uterine tissues.
  • The term “antagonist” as used herein, refers to a small-molecule agent that binds to a nuclear hormone receptor and subsequently decreases the agonist induced transcriptional activity of the nuclear hormone receptor.
  • The term “agonist” as used herein, refers to a small-molecule agent that binds to a nuclear hormone receptor and subsequently increases nuclear hormone receptor transcriptional activity in the absence of a known agonist.
  • The term “inverse agonist” as used herein, refers to a small-molecule agent that binds to a nuclear hormone receptor and subsequently decreases the basal level of nuclear hormone receptor transcriptional activity that is present in the absence of a known agonist.
  • The term “degrader” as used herein, refers to a small molecule agent that binds to a nuclear hormone receptor and subsequently lowers the steady state protein levels of said receptor. In some embodiments, a degrader as described herein lowers steady state estrogen receptor levels by at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90% or at least 95%.
  • The term “selective estrogen receptor degrader” or “SERD” as used herein, refers to a small molecule agent that preferentially binds to estrogen receptors versus other receptors and subsequently lowers the steady state estrogen receptor levels.
  • The term “ER-dependent”, as used herein, refers to diseases or conditions that would not occur, or would not occur to the same extent, in the absence of estrogen receptors.
  • The term “ER-mediated”, as used herein, refers to diseases or conditions that occur in the absence of estrogen receptors but can occur in the presence of estrogen receptors.
  • The term “ER-sensitive”, as used herein, refers to diseases or conditions that would not occur, or would not occur to the same extent, in the absence of estrogens.
  • The term “cancer” as used herein refers to an abnormal growth of cells which tend to proliferate in an uncontrolled way and, in some cases, to metastasize (spread). The types of cancer include, but is not limited to, solid tumors (such as those of the bladder, bowel, brain, breast, endometrium, heart, kidney, lung, uterus, lymphatic tissue (lymphoma), ovary, pancreas or other endocrine organ (thyroid), prostate, skin (melanoma or basal cell cancer) or hematological tumors (such as the leukemias and lymphomas) at any stage of the disease with or without metastases.
  • Additional non-limiting examples of cancers include, acute lymphoblastic leukemia, acute myeloid leukemia, adrenocortical carcinoma, anal cancer, appendix cancer, astrocytomas, atypical teratoid/rhabdoid tumor, basal cell carcinoma, bile duct cancer, bladder cancer, bone cancer (osteosarcoma and malignant fibrous histiocytoma), brain stem glioma, brain tumors, brain and spinal cord tumors, breast cancer, bronchial tumors, Burkitt lymphoma, cervical cancer, chronic lymphocytic leukemia, chronic myelogenous leukemia, colon cancer, colorectal cancer, craniopharyngioma, cutaneous T-Cell lymphoma, embryonal tumors, endometrial cancer, ependymoblastoma, ependymoma, esophageal cancer, ewing sarcoma family of tumors, eye cancer, retinoblastoma, gallbladder cancer, gastric (stomach) cancer, gastrointestinal carcinoid tumor, gastrointestinal stromal tumor (GIST), gastrointestinal stromal cell tumor, germ cell tumor, glioma, hairy cell leukemia, head and neck cancer, hepatocellular (liver) cancer, hodgkin lymphoma, hypopharyngeal cancer, intraocular melanoma, islet cell tumors (endocrine pancreas), Kaposi sarcoma, kidney cancer, Langerhans cell histiocytosis, laryngeal cancer, leukemia, Acute lymphoblastic leukemia, acute myeloid leukemia, chronic lymphocytic leukemia, chronic myelogenous leukemia, hairy cell leukemia, liver cancer, non-small cell lung cancer, small cell lung cancer, Burkitt lymphoma, cutaneous T-cell lymphoma, Hodgkin lymphoma, non-Hodgkin lymphoma, lymphoma, Waldenström macroglobulinemia, medulloblastoma, medulloepithelioma, melanoma, mesothelioma, mouth cancer, chronic myelogenous leukemia, myeloid leukemia, multiple myeloma, nasopharyngeal cancer, neuroblastoma, non-Hodgkin lymphoma, non-small cell lung cancer, oral cancer, oropharyngeal cancer, osteosarcoma, malignant fibrous histiocytoma of bone, ovarian cancer, ovarian epithelial cancer, ovarian germ cell tumor, ovarian low malignant potential tumor, pancreatic cancer, papillomatosis, parathyroid cancer, penile cancer, pharyngeal cancer, pineal parenchymal tumors of intermediate differentiation, pineoblastoma and supratentorial primitive neuroectodermal tumors, pituitary tumor, plasma cell neoplasm/multiple myeloma, pleuropulmonary blastoma, primary central nervous system lymphoma, prostate cancer, rectal cancer, renal cell (kidney) cancer, retinoblastoma, rhabdomyosarcoma, salivary gland cancer, sarcoma, Ewing sarcoma family of tumors, sarcoma, kaposi, Sézary syndrome, skin cancer, small cell Lung cancer, small intestine cancer, soft tissue sarcoma, squamous cell carcinoma, stomach (gastric) cancer, supratentorial primitive neuroectodermal tumors, T-cell lymphoma, testicular cancer, throat cancer, thymoma and thymic carcinoma, thyroid cancer, urethral cancer, uterine cancer, uterine sarcoma, vaginal cancer, vulvar cancer, Waldenström macroglobulinemia, Wilms tumor.
  • The terms “co-administration” or the like, as used herein, are meant to encompass administration of the selected therapeutic agents to a single patient, and are intended to include treatment regimens in which the agents are administered by the same or different route of administration or at the same or different time.
  • The terms “effective amount” or “therapeutically effective amount,” as used herein, refer to a sufficient amount of an agent or a compound being administered which will relieve to some extent one or more of the symptoms of the disease or condition being treated. The result includes reduction and/or alleviation of the signs, symptoms, or causes of a disease, or any other desired alteration of a biological system. For example, an “effective amount” for therapeutic uses is the amount of the composition comprising a compound as disclosed herein required to provide a clinically significant decrease in disease symptoms. An appropriate “effective” amount in any individual case is optionally determined using techniques, such as a dose escalation study.
  • The term “pharmaceutical combination” as used herein, means a product that results from the mixing or combining of more than one active ingredient and includes both fixed and non-fixed combinations of the active ingredients. The term “fixed combination” means that the active ingredients, e.g. a compound of Formula (I), and a co-agent, are both administered to a patient simultaneously in the form of a single entity or dosage. The term “non-fixed combination” means that the active ingredients, e.g. a compound of Formula (I), and a co-agent, are administered to a patient as separate entities either simultaneously, concurrently or sequentially with no specific intervening time limits, wherein such administration provides effective levels of the two compounds in the body of the patient. The latter also applies to cocktail therapy, e.g. the administration of three or more active ingredients.
  • A “metabolite” of a compound disclosed herein is a derivative of that compound that is formed when the compound is metabolized. The term “active metabolite” refers to a biologically active derivative of a compound that is formed when the compound is metabolized. The term “metabolized,” as used herein, refers to the sum of the processes (including, but not limited to, hydrolysis reactions and reactions catalyzed by enzymes) by which a particular substance is changed by an organism. Thus, enzymes may produce specific structural alterations to a compound. For example, cytochrome P450 catalyzes a variety of oxidative and reductive reactions while uridine diphosphate glucuronyltransferases catalyze the transfer of an activated glucuronic-acid molecule to aromatic alcohols, aliphatic alcohols, carboxylic acids, amines and free sulphydryl groups. Metabolites of the compounds disclosed herein are optionally identified either by administration of compounds to a host and analysis of tissue samples from the host, or by incubation of compounds with hepatic cells in vitro and analysis of the resulting compounds.
  • The term “subject” or “patient” encompasses mammals. Examples of mammals include, but are not limited to humans, chimpanzees, apes, monkeys, cattle, horses, sheep, goats, swine, rabbits, dogs, cats, rats, mice, guinea pigs, and the like. In one aspect, the mammal is a human.
  • The terms “treat,” “treating” or “treatment,” as used herein, include alleviating, abating or ameliorating at least one symptom of a disease or condition, preventing additional symptoms, inhibiting the disease or condition, e.g., arresting the development of the disease or condition, relieving the disease or condition, causing regression of the disease or condition, relieving a condition caused by the disease or condition, or stopping the symptoms of the disease or condition either prophylactically and/or therapeutically.
  • Routes of Administration
  • Suitable routes of administration include, but are not limited to, oral, parenteral (e.g., intravenous, subcutaneous, intramuscular), intranasal, buccal, topical, rectal, aerosol, ophthalmic, pulmonary, transmucosal, transdermal, vaginal, otic, nasal, and topical administration. In addition, by way of example only, parenteral delivery includes intramuscular, subcutaneous, intravenous, intramedullary injections, as well as intrathecal, direct intraventricular, intraperitoneal, intralymphatic, and intranasal injections. In certain embodiments, a compound as described herein is administered in a systemic manner. In certain other embodiments, a compound as described herein is administered in a local rather than systemic manner.
  • Pharmaceutical Compositions/Formulations
  • In some embodiments, the compounds described herein are formulated into pharmaceutical compositions. Pharmaceutical compositions are formulated in a conventional manner using one or more pharmaceutically acceptable inactive ingredients that facilitate processing of the active compounds into preparations that are used pharmaceutically. Proper formulation is dependent upon the route of administration chosen. A summary of pharmaceutical compositions described herein is found, for example, in Remington: The Science and Practice of Pharmacy, Nineteenth Ed (Easton, Pa.: Mack Publishing Company, 1995); Hoover, John E., Remington's Pharmaceutical Sciences, Mack Publishing Co., Easton, Pa. 1975; Liberman, H. A. and Lachman, L., Eds., Pharmaceutical Dosage Forms, Marcel Decker, New York, N.Y., 1980; and Pharmaceutical Dosage Forms and Drug Delivery Systems, Seventh Ed. (Lippincott Williams & Wilkins 1999), herein incorporated by reference for such disclosure.
  • A pharmaceutical composition, as used herein, refers to a mixture of a compound of Formula (I), with other chemical components (i.e. pharmaceutically acceptable inactive ingredients), such as carriers, excipients, binders, filling agents, suspending agents, flavoring agents, sweetening agents, disintegrating agents, dispersing agents, surfactants, lubricants, colorants, diluents, solubilizers, moistening agents, plasticizers, stabilizers, penetration enhancers, wetting agents, anti-foaming agents, antioxidants, preservatives, or one or more combination thereof. The pharmaceutical composition facilitates administration of the compound to a mammal.
  • The pharmaceutical compositions will include at least one compound of Formula (I), or a pharmaceutically acceptable salt thereof, as an active ingredient in free-acid or free-base form, or in a pharmaceutically acceptable salt form. In addition, the methods and pharmaceutical compositions described herein include the use of N-oxides (if appropriate), crystalline forms, amorphous phases, as well as active metabolites of these compounds having the same type of activity. In some embodiments, compounds described herein exist in unsolvated form or in solvated forms with pharmaceutically acceptable solvents such as water, ethanol, and the like.
  • The pharmaceutical formulations described herein include, but are not limited to, aqueous liquid dispersions, self-emulsifying dispersions, solid solutions, liposomal dispersions, aerosols, solid dosage forms, powders, immediate release formulations, controlled release formulations, fast melt formulations, tablets, capsules, pills, delayed release formulations, extended release formulations, enteric coated formulations, pulsatile release formulations, multiparticulate formulations, and mixed immediate and controlled release formulations.
  • In some embodiments, the compound of Formula (I), or a pharmaceutically acceptable salt thereof, is administered systemically.
  • In some embodiments, the compound of Formula (I), or a pharmaceutically acceptable salt thereof, is administered orally. All formulations for oral administration are in dosages suitable for such administration. In some embodiments, the solid dosage forms disclosed herein are in the form of a tablet, a pill, a powder, a capsule, solid dispersion, solid solution, bioerodible dosage form, controlled release formulations, pulsatile release dosage forms, multiparticulate dosage forms, beads, pellets, granules. In other embodiments, the pharmaceutical formulation is in the form of a powder. In still other embodiments, the pharmaceutical formulation is in the form of a tablet. In still other embodiments, the pharmaceutical formulation is in the form of a suspension tablet, a fast-melt tablet, a bite-disintegration tablet, a rapid-disintegration tablet, an effervescent tablet, or a caplet. In other embodiments, pharmaceutical formulation is in the form of a capsule.
  • In some embodiments, the pharmaceutical solid oral dosage forms are formulated to provide a controlled release of the active compound. Controlled release profiles include, for example, sustained release, prolonged release, pulsatile release, and delayed release profiles.
  • In one aspect, liquid formulation dosage forms for oral administration are in the form of aqueous suspensions selected from the group including, but not limited to, pharmaceutically acceptable aqueous oral dispersions, emulsions, solutions, elixirs, gels, and syrups. See, e.g., Singh et al., Encyclopedia of Pharmaceutical Technology, 2nd Ed., pp. 754-757 (2002).
  • For buccal or sublingual administration, the compositions optionally take the form of tablets, lozenges, or gels formulated in a conventional manner.
  • In one aspect, a compound of Formula (I), or a pharmaceutically acceptable salt thereof, is formulated into a pharmaceutical composition suitable for intramuscular, subcutaneous, or intravenous injection. Parenteral injections involve either bolus injection and/or continuous infusion.
  • In some embodiments, the compound of Formula (I), or a pharmaceutically acceptable salt thereof, is administered intravenously. In some embodiments, the compound of Formula (I), or a pharmaceutically acceptable salt thereof, is administered subcutaneously.
  • In some embodiments, the compound of Formula (I), or a pharmaceutically acceptable salt thereof, is administered topically. In such embodiments, the compound of Formula (I), or a pharmaceutically acceptable salt thereof, is formulated into a variety of topically administrable compositions, such as solutions, suspensions, lotions, gels, pastes, shampoos, scrubs, rubs, smears, medicated sticks, medicated bandages, balms, creams or ointments. In some embodiments, the compound of Formula (I), or a pharmaceutically acceptable salt thereof, is administered topically to the skin of mammal. In some embodiments, the compound of Formula (I), is prepared as a transdermal dosage form.
  • In another aspect is the use of a compound of Formula (I), or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for treating a disease, disorder or conditions in which the activity of estrogen receptors contributes to the pathology and/or symptoms of the disease or condition. In one aspect, the disease or condition is any of the diseases or conditions specified herein.
  • Methods of Dosing and Treatment Regimens
  • In one embodiment, the compound of Formula (I), or a pharmaceutically acceptable salt thereof, is used in the preparation of medicaments for the treatment of diseases or conditions in a mammal that would benefit from a reduction of estrogen receptor activity. Methods for treating any of the diseases or conditions described herein in a mammal in need of such treatment, involves administration of pharmaceutical compositions that include at least one compound of Formula (I), or a pharmaceutically acceptable salt, N-oxide, active metabolite, prodrug, or pharmaceutically acceptable solvate thereof, in therapeutically effective amounts to said mammal.
  • Therapeutically effective amounts depend on the severity and course of the disease or condition, previous therapy, the patient's health status, weight, and response to the drugs, and the judgment of the treating physician. Therapeutically effective amounts are optionally determined by methods including, but not limited to, a dose escalation clinical trial.
  • In any of the method of treatments described herein, the effective amount of the compound of Formula (I) is: (a) systemically administered to the mammal; and/or (b) administered orally to the mammal; and/or (c) intravenously administered to the mammal; and/or (d) administered by injection to the mammal; and/or (e) administered topically to the mammal; and/or (f) administered non-systemically or locally to the mammal.
  • In some situations the methods of treatment comprise single administration of the effective amount of the compound, including further embodiments in which (i) the compound is administered once; (ii) the compound is administered to the mammal multiple times over the span of one day; (iii) continually; or (iv) continuously.
  • In any of the aforementioned aspects are further embodiments comprising multiple administrations of the effective amount of the compound, including further embodiments in which (i) the compound is administered continuously or intermittently: as in a single dose; (ii) the time between multiple administrations is every 6 hours; (iii) the compound is administered to the mammal every 8 hours; (iv) the compound is administered to the mammal every 12 hours; (v) the compound is administered to the mammal every 24 hours. In further or alternative embodiments, the method comprises a drug holiday, wherein the administration of the compound is temporarily suspended or the dose of the compound being administered is temporarily reduced; at the end of the drug holiday, dosing of the compound is resumed. In one embodiment, the length of the drug holiday varies from 2 days to 1 year.
  • In certain embodiments wherein the patient's condition does not improve, upon the doctor's discretion the compound is administered chronically, that is, for an extended period of time.
  • In certain embodiments wherein a patient's status does improve, the dose of drug being administered is temporarily reduced or temporarily suspended for a certain length of time (i.e., a “drug holiday”).
  • In some embodiments, doses employed for adult human treatment are typically in the range of 0.01 mg-5000 mg per day. In one aspect, doses employed for adult human treatment are from about 1 mg to about 1000 mg per day. In one embodiment, the desired dose is conveniently presented in a single dose or in divided doses administered simultaneously or at appropriate intervals, for example as two, three, four or more sub-doses per day. In one embodiment, the daily dosages appropriate for the compound of Formula (I), or a pharmaceutically acceptable salt thereof, described herein are from about 0.01 to about 50 mg/kg per body weight.
  • Combination Therapy
  • In certain instances, it is appropriate to administer at least one compound of Formula (I), or a pharmaceutically acceptable salt thereof, in combination with one or more other therapeutic agents. In certain embodiments, the pharmaceutical composition further comprises one or more anti-cancer agents.
  • In one specific embodiment, a compound of Formula (I), or a pharmaceutically acceptable salt thereof, is co-administered with a second therapeutic agent, wherein the compound of Formula (I), or a pharmaceutically acceptable salt thereof, and the second therapeutic agent modulate different aspects of the disease, disorder or condition being treated, thereby providing a greater overall benefit than administration of either therapeutic agent alone.
  • In any case, regardless of the disease, disorder or condition being treated, the overall benefit experienced by the patient is the additive effect of the two therapeutic agents or the patient may experience a synergistic benefit.
  • In combination therapies, the multiple therapeutic agents (one of which is one of the compounds described herein) are administered in any order or even simultaneously. If administration is simultaneous, the multiple therapeutic agents are, by way of example only, provided in a single, unified form, or in multiple forms (e.g., as a single pill or as two separate pills).
  • In some embodiments, methods for treatment of estrogen receptor-dependent or estrogen receptor-mediated conditions or diseases, such as proliferative disorders, including cancer, comprises administration to a mammal a compound of Formula (I), or a pharmaceutically acceptable salt thereof, in combination with at least one additional therapeutic agent.
  • In some embodiments, a compound of Formula (I), or a pharmaceutically acceptable salt thereof, in combination with hormone blocking therapy, chemotherapy, radiation therapy, monoclonal antibodies, or combinations thereof.
  • In some embodiments, the at least one additional therapeutic agent for use in combination with a compound of Formula (I), or a pharmaceutically acceptable salt thereof, include one or more of the following: abiraterone; abarelix; adriamycin; actinomycin; acivicin; aclarubicin; acodazole hydrochloride; acronine; adozelesin; aldesleukin; alemtuzumab; allopurinol; alitretinoin; altretamine; ambomycin; ametantrone acetate; aminoglutethimide; aminolevulinic acid; amifostine; amsacrine; anastrozole; anthramycin; aprepitant; arsenic trioxide; asparaginase; asperlin; azacitidine; azetepa; azotomycin; batimastat; bendamustine hydrochloride; benzodepa; bevacizumab; bexarotene; bicalutamide; bisantrene hydrochloride; bisnafide dimesylate; bizelesin; bleomycin; bleomycin sulfate; bortezomib; brequinar sodium; bropirimine; busulfan; cactinomycin; calusterone; caracemide; carbetimer; carboplatin; carmustine; carubicin hydrochloride; carzelesin; capecitabine; cedefingol; cetuximab; chlorambucil; cirolemycin; cisplatin; cladribine; clofarabine; crisnatol mesylate; cyclophosphamide; cytarabine; dacarbazine; dasatinib; daunorubicin hydrochloride; dactinomycin; darbepoetin alfa; decitabine; degarelix; denileukin diftitox; dexormaplatin; dexrazoxane hydrochloride; dezaguanine; dezaguanine mesylate; diaziquone; docetaxel; doxorubicin; doxorubicin hydrochloride; droloxifene; droloxifene citrate; dromostanolone propionate; duazomycin; edatrexate; eflornithine hydrochloride; elsamitrucin; eltrombopag olamine; enloplatin; enpromate; epipropidine; epirubicin hydrochloride; epoetin alfa; erbulozole; erlotinib hydrochloride; esorubicin hydrochloride; estramustine; estramustine phosphate sodium; etanidazole; etoposide; etoposide phosphate; etoprine; everolimus; exemestane; fadrozole hydrochloride; fazarabine; fenretinide; filgrastim; floxuridine; fludarabine phosphate; fluorouracil; fluorocitabine; fosquidone; fostriecin sodium; fulvestrant; gefitinib; gemcitabine; gemcitabine hydrochloride; gemcitabine-cisplatin; gemtuzumab ozogamicin; goserelin acetate; histrelin acetate; hydroxyurea; idarubicin hydrochloride; ifosfamide; iimofosine; ibritumomab tiuxetan; idarubicin; ifosfamide; imatinib mesylate; imiquimod; interleukin II (including recombinant interleukin II, or r1L2), interferon alfa-2a; interferon alfa-2b; interferon alfa-n1; interferon alfa-n3; interferon beta-1a; interferon gamma-1b; iproplatin; irinotecan hydrochloride; ixabepilone; lanreotide acetate; lapatinib; lenalidomide; letrozole; leuprolide acetate; leucovorin calcium; leuprolide acetate; levamisole; liposomal cytarabine; liarozole hydrochloride; lometrexol sodium; lomustine; losoxantrone hydrochloride; masoprocol; maytansine; mechlorethamine hydrochloride; megestrol acetate; melengestrol acetate; melphalan; menogaril; mercaptopurine; methotrexate; methotrexate sodium; methoxsalen; metoprine; meturedepa; mitindomide; mitocarcin; mitocromin; mitogillin; mitomalcin; mitomycin C; mitosper; mitotane; mitoxantrone hydrochloride; mycophenolic acid; nandrolone phenpropionate; nelarabine; nilotinib; nocodazoie; nofetumomab; nogalamycin; ofatumumab; oprelvekin; ormaplatin; oxaliplatin; oxisuran; paclitaxel; palifermin; palonosetron hydrochloride; pamidronate; pegfilgrastim; pemetrexed disodium; pentostatin; panitumumab; pazopanib hydrochloride; pemetrexed disodium; plerixafor; pralatrexate; pegaspargase; peliomycin; pentamustine; peplomycin sulfate; perfosfamide; pipobroman; piposulfan; piroxantrone hydrochloride; plicamycin; plomestane; porfimer sodium; porfiromycin; prednimustine; procarbazine hydrochloride; puromycin; puromycin hydrochloride; pyrazofurin; quinacrine; raloxifene hydrochloride; rasburicase; recombinant HPV bivalent vaccine; recombinant HPV quadrivalent vaccine; riboprine; rogletimide; rituximab; romidepsin; romiplostim; safingol; safingol hydrochloride; sargramostim; semustine; simtrazene; sipuleucel-T; sorafenib; sparfosate sodium; sparsomycin; spirogermanium hydrochloride; spiromustine; spiroplatin; streptonigrin; streptozocin; sulofenur; sunitinib malate; talisomycin; tamoxifen citrate; tecogalan sodium; tegafur; teloxantrone hydrochloride; temozolomide; temoporfin; temsirolimus; teniposide; teroxirone; testolactone; thalidomide; thiamiprine; thioguanine; thiotepa; tiazofurin; tirapazamine; topotecan hydrochloride; toremifene; tositumomab and I 131 Iodine tositumomab; trastuzumab; trestolone acetate; tretinoin; triciribine phosphate; trimetrexate; trimetrexate glucuronate; triptorelin; tubulozole hydrochloride; uracil mustard; uredepa; valrubicin; vapreotide; verteporfin; vinblastine; vinblastine sulfate; vincristine sulfate; vindesine; vindesine sulfate; vinepidine sulfate; vinglycinate sulfate; vinleurosine sulfate; vinorelbine tartrate; vinrosidine sulfate; vinzolidine sulfate; vorinostat; vorozole; zeniplatin; zinostatin; zoledronic acid; and zorubicin hydrochloride.
  • In some embodiments, a compound of Formula (I), or a pharmaceutically acceptable salt thereof, is used in combination with anti-emetic agents to treat nausea or emesis, which result from the use of a compound of Formula (I), anti-cancer agent(s) and/or radiation therapy.
  • In some embodiments, a compound of Formula (I), or a pharmaceutically acceptable salt thereof, is used in combination with an agent useful in the treatment of anemia or neutropenia.
  • In some embodiments, a compound of Formula (I), or a pharmaceutically acceptable salt thereof, is administered with corticosteroids.
  • In some embodiments, a compound of Formula (I), or a pharmaceutically acceptable salt thereof, is coadministered with analgesics.
  • In some embodiments, a compound of Formula (I), or a pharmaceutically acceptable salt thereof, is used in combination with radiation therapy (or radiotherapy). Radiation therapy is the treatment of cancer and other diseases with ionizing radiation. Radiation therapy is optionally used to treat localized solid tumors, such as cancers of the skin, tongue, larynx, brain, breast, prostate, colon, uterus and/or cervix. It is also optionally used to treat leukemia and lymphoma (cancers of the blood-forming cells and lymphatic system, respectively).
  • A technique for delivering radiation to cancer cells is to place radioactive implants directly in a tumor or body cavity. This is called internal radiotherapy (brachytherapy, interstitial irradiation, and intracavitary irradiation are types of internal radiotherapy.) Using internal radiotherapy, the radiation dose is concentrated in a small area, and the patient stays in the hospital for a few days. Internal radiotherapy is frequently used for cancers of the tongue, uterus, prostate, colon, and cervix. The term “radiotherapy” or “ionizing radiation” include all forms of radiation, including but not limited to α, β, and γ radiation and ultraviolet light.
  • EXAMPLES
  • These examples are provided for illustrative purposes only and not to limit the scope of the claims provided herein.
  • Example 1 Preparation of 5-Bromo-1-(tetrahydro-2H-pyran-2-yl)-1H-indazole (Intermediate 1)
  • Figure US20120071535A1-20120322-C00158
  • A 250-mL round-bottom flask equipped with a magnetic stir bar, a rubber septum, and a N2 inlet was charged with 5-bromo-1H-indazole (10 g, 50.7 mmol) and anhydrous DCM (101 mL). To this solution, DHP (23 mL, 253.8 mmol) was added in one portion at room temperature followed by addition of PPTS (1.28 g, 5 mmol). The resulting mixture was stirred at room temperature for 48 h. Upon completion by TLC, the reaction mixture was quenched with water and extracted with DCM (3×100 mL). The combined organic extracts were washed with water (100 mL), washed with brine (50 mL), dried over sodium sulfate, filtered, concentrated, and purified by silica gel chromatography (0-10% ethyl acetate in hexanes) to give the title compound (13 g) as a pale yellow oil. 1H NMR (300 MHz, DMSO-d6): δ 8.10 (s, 1H), 8.02 (d, 1H), 7.73 (d, 1H), 7.53 (dd, 1H), 5.86 (dd, 1H), 3.89-3.85 (m, 1H), 3.73-3.69 (m, 1H), 2.43-2.31 (m, 1H), 2.06-1.92 (m, 2H), 1.80-1.64 (m, 1H), 1.60-150 (m, 2H).
  • Example 2 Preparation of 5-Ethynyl-1-(tetrahydro-2H-pyran-2-yl)-1H-indazole (Intermediate 2) Step 1: 1-(Tetrahydro-2H-pyran-2-yl)-5-((trimethylsilyl)ethynyl)-1H-indazole
  • Figure US20120071535A1-20120322-C00159
  • To a 250-mL pressure tube, 5-bromo-1-(tetrahydro-2H-pyran-2-yl)-1H-indazole (11.9 g, 42.3 mmol; Intermediate 1), Pd(Ph3P)2Cl2 (1.48 g, 0.05 mmol), CuI (0.8 g, 4.2 mmol) and THF/triethylamine (5:1, 85 mL) were added. This mixture was degassed with three vacuum/N2 cycles, and then trimethylsilylacetylene (9 mL, 63.5 mmol) was added. The pressure tube was sealed and heated at 80° C. for 2 days. Upon completion by LCMS, the reaction mixture was cooled down to room temperature and filtered through Celite with ethyl acetate (200 mL). The filtrate was concentrated to give the crude product that was used directly in the next step. LCMS: 299 (M+H)+.
  • Note: For this compound and other compounds synthesized using this reaction, alternate procedures have been employed using an amine, such as triethylamine or pyrrolidine, as the sole solvent.
  • Step 2: 5-Ethynyl-1-(tetrahydro-2,1-pyran-2-yl)-1H-indazole
  • Figure US20120071535A1-20120322-C00160
  • A 250-mL round-bottom flask equipped with a magnetic stir bar, a rubber septum, and a N2 inlet was charged with a solution of 1-(tetrahydro-2H-pyran-2-yl)-5-((trimethylsilypethynyl)-1H-indazole (12.6 g, 42.2 mmol) in MeOH. To this solution, solid K2CO3 (0.58 g, 4.2 mmol) was added in one portion. The resulting mixture was stirred at room temperature for 4 h. Upon completion by TLC, the reaction mixture was filtered, concentrated, and purified by silica gel chromatography (0-10% ethyl acetate in hexanes) to give the title compound (4.7 g) as a pale yellow solid. 1H NMR (300 MHz, DMSO-d6): δ 8.13 (s, 1H), 7.96 (s, 1H), 7.75 (d, 1H), 7.47 (dd, 1H), 5.86 (dd, 1H), 4.10 (s, 1H), 3.90-3.86 (m, 1H), 3.78-3.68 (m, 1H), 2.43-2.32 (m, 1H), 2.06-1.93 (m, 2H), 1.81-1.66 (m, 1H), 1.60-1.50 (m, 2H).
  • Example 3 Preparation of 5-(But-1-yn-1-yl)-1-(tetrahydro-2H-pyran-2-yl)-1H-indazole (Intermediate 3)
  • Figure US20120071535A1-20120322-C00161
  • A 250-mL round-bottom flask equipped with a magnetic stir bar, a rubber septum, and a N2 inlet was charged with 5-ethynyl-1-(tetrahydro-2H-pyran-2-yl)-1H-indazole (4.2 g, 18.6 mmol; Intermediate 2) and anhydrous THF/TMEDA (9:1, 93 mL). This solution was cooled to −78° C. in an IPA/dry ice bath, and n-BuLi (17.4 mL solution in hexanes, 27.84 mmol) was added dropwise over 15 minutes. The resulting mixture was stirred for 30 minutes at −78° C., and then iodoethane (2.23 mL, 27.84 mmol) was added dropwise over 5 minutes. The mixture was gradually warmed to room temperature, stirred for 1 h, and then heated at 40° C. overnight. Upon completion by LCMS, the reaction mixture was cooled to room temperature, quenched with water (100 mL), and extracted with ethyl acetate (2×100 mL). The combined organics were washed with water (100 mL), washed with brine (50 mL), dried over sodium sulfate, filtered, concentrated, and purified by silica gel chromatography (0-10% ethyl acetate in hexanes) to give the title compound (1.42 g) as a pale yellow solid. 1H NMR (300 MHz, DMSO-d6): δ 8.08 (s, 1H), 7.82 (s, 1H), 7.69 (d, 1H), 7.39 (d, 1H), 5.84 (dd, 1H), 3.89-3.86 (m, 1H), 3.76-3.72 (m, 1H), 2.45-2.36 (m, 3H), 2.04-1.94 (m, 2H), 1.74 (m, 1H), 1.57-1.20 (m, 2H), 1.16 (t, 3H); LCMS: 255 (M+H)+.
  • Note: For this compound and other compounds prepared using this reaction, lithium bis(trimethylsilyl)amide has been employed as the base in THF at 0° C. followed by alkylation with alkyl-halide at reflux.
  • The Intermediates in Table 2 were prepared from known or commercial starting materials following the procedures outlined for Intermediates 1-3.
  • TABLE 2
    Intermediate 4 1-(Tetrahydro-2H-pyran-2-yl)-5-(4,4,4- trideuterobut-1-yn-1-yl)-1H-indazole
    Figure US20120071535A1-20120322-C00162
    Intermediate 5 5-(But-1-yn-1-yl)-7-chloro-1-(tetrahydro-2H- pyran-2-yl)-1H-indazole
    Figure US20120071535A1-20120322-C00163
    Intermediate 6 5-(But-1-yn-1-yl)-7-methyl-1-(tetrahydro-2H- pyran-2-yl)-1H-indazole
    Figure US20120071535A1-20120322-C00164
    Intermediate 7 5-(But-1-yn-1-yl)-3-methyl-1-(tetrahydro-2H- pyran-2-yl)-1H-indazole
    Figure US20120071535A1-20120322-C00165
    Intermediate 8 5-(But-1-yn-1-yl)-3-chloro-1-(tetrahydro-2H- pyran-2-yl)-1H-indazole
    Figure US20120071535A1-20120322-C00166
    Intermediate 9 5-(Prop-1-yn-1-yl)-1-(tetrahydro-2H-pyran-2-yl)- 1H-indazole
    Figure US20120071535A1-20120322-C00167
    Intermediate 10 5-(Pent-1-yn-1-yl)-1-(tetrahydro-2H-pyran-2-yl)- 1H-indazole
    Figure US20120071535A1-20120322-C00168
    Intermediate 11 5-(Perdeuterobut-1-yn-1-yl)-1-(tetrahydro-2H- pyran-2-yl)-1H-indazole
    Figure US20120071535A1-20120322-C00169
  • Example 4 Preparation of 5-(But-1-yn-1-yl)-4-methyl-1-(tetrahydro-2H-pyran-2-yl)-1H-indazole (Intermediate 12) Step 1: 4-Methyl-1-(tetrahydro-2H-pyran-2-yl)-5-((trimethylsilyl)ethynyl)-1H-indazole
  • Figure US20120071535A1-20120322-C00170
  • A mixture of 5-bromo-4-methyl-1-(tetrahydro-2H-pyran-2-yl)-1H-indazole (915 mg, 3.10 mmol; prepared from 5-bromo-4-methyl-1H-indazole following the procedure outlined for Intermediate 1), copper iodide (72 mg, 0.38 mmol), sodium tetrachloropalladate (55 mg, 0.19 mmol), 2-(di-tert-butylphosphino)-1-phenyl-1H-indole (128 mg, 0.379 mmol), and TMEDA:H2O (9:1, 10 mL) was degassed with three vacuum/nitrogen cycles. Ethynyltrimethylsilane was added to the reaction, and the mixture was heated at 80° C. for 90 min and then cooled to room temperature. The reaction mixture was filtered through Celite and the Celite was washed with ethyl acetate (100 mL). The filtrate was washed (2×50 mL sat'd NaHCO3), dried (Na2SO4), and concentrated under reduced pressure. The crude material was purified on a silica gel column to yield the desired compound. 1H NMR (300 MHz, DMSO-d6): δ 8.24 (s, 1H), 7.54 (d, 1H), 7.39 (d, 1H), 5.82 (dd, 1H), 3.88 (m, 1H), 3.71 (m, 1H), 2.63 (s, 3H), 2.39 (m, 1H), 2.00 (m, 2H), 1.72 (m, 1H), 1.58 (m, 2H), 0.24 (s, 9H); LCMS: 313 (M+H)+.
  • Step 2: 5-(But-1-yn-1-yl)-4-methyl-1-(tetrahydro-2H-pyran-2-yl)-1H-indazole
  • Figure US20120071535A1-20120322-C00171
  • The title compound was prepared from 4-methyl-1-(tetrahydro-2H-pyran-2-yl)-5-((trimethylsilypethynyl)-1H-indazole following the procedures outlined for Intermediate 2 (step 2) and Intermediate 3. 1H NMR (300 MHz, DMSO-d6): δ 7.97 (s, 1H), 7.27 (d, 1H), 7.11 (d, 1H), 5.59 (dd, 1H), 3.58 (m, 1H), 3.50 (m, 1H), 2.38 (s, 3H), 2.17 (q, 2H), 2.13 (m, 1H), 1.77 (m, 2H), 1.50 (m, 1H), 1.36 (m, 2H), 0.98 (t, 3H).
  • The Intermediate in Table 3 was prepared from 5-bromo-6-methyl-1H-indazole following the procedures outlined for Intermediate 12.
  • TABLE 3
    Intermediate 13 5-(But-1-yn-1-yl)-6-methyl-1-(tetrahydro- 2H-pyran-2-yl)-1H-indazole
    Figure US20120071535A1-20120322-C00172
  • Example 5 Preparation of 5-(Cyclopropylethynyl)-1-(tetrahydro-2H-pyran-2-yl)-1H-indazole (Intermediate 14)
  • Figure US20120071535A1-20120322-C00173
  • A 1 L three-necked round bottom flask was charged with 5-bromo-1-(tetrahydro-2H-pyran-2-yl)-1H-indazole (31.2 g, 111 mmol; Intermediate 1) and triethylamine (500 mL). The flask was degassed with three vacuum/N2 cycles, followed by the addition of Pd(PPh3)2Cl2 (7.7 g, 11 mmol) and CuI (2.1 g, 11 mmol) under N2 atmosphere. The flask was again degassed with three vacuum/N2 cycles. Ethynylcyclopropane (70% in toluene, 20.9 g, 222 mmol) was then added via syringe and the reaction mixture was stirred at 80° C. for 16 hours. Upon completion, the solvent was evaporated. The residue was diluted with dichloromethane (600 mL), washed with water (2×200 mL) and brine (200 mL), dried over anhydrous sodium sulfate, and concentrated in vacuo. The residue was further purified on silica gel column (1:100-1:20 EtOAc/petroleum ether) affording the title compound (27.0 g). 1H NMR (400 MHz, DMSO-d6): δ 8.09 (s, 1H), 7.82 (s, 1H), 7.70 (d, 1H), 7.39 (m, 1H), 5.84 (dd, 1H), 3.91-3.87 (m, 1H), 3.78-3.73 (m, 1H), 2.52-2.37 (m, 1H), 2.05-1.94 (m, 2H), 1.76-1.72 (m, 1H), 1.60-1.52 (m, 3H), 0.92-0.87 (m, 2H), 0.78-0.73 (m, 2H); LCMS: 267 (M+H)±.
  • Example 6 Preparation of 5-(4-Methylpent-1-yn-1-yl)-1-(tetrahydro-2H-pyran-2-yl)-1H-indazole (Intermediate 15)
  • Figure US20120071535A1-20120322-C00174
  • To a mixture of 5-bromo-1-(tetrahydro-2H-pyran-2-yl)-1H-indazole (3.0 g, 10.7 mmol; Intermediate 1), Pd(PPh3)2Cl2 (1.03 g, 1.07 mmol) and CuI (203 mg, 1.07 mmol) in triethylamine (30 mL), was added 4-methylpent-1-yne (2.23 g, 27.8 mmol) under N2 atmosphere. The resulting mixture was stirred at 80° C. for 16 hours under N2 atmosphere. Upon completion, the reaction mixture was diluted with EtOAc and filtered. The filtrate was washed with water (3×10 mL), dried over Na2SO4 and concentrated in vacuo. The residue was purified by column chromatography on silica gel (0-10% EtOAc in petroleum ether) affording the title compound (2.2 g) as yellow solid. 1H NMR (400 MHz, DMSO-d6): δ 8.09 (s, 1H), 7.84 (s, 1H), 7.69 (d, 1H), 7.39 (dd, 1H), 5.83 (dd, 1H), 3.90-3.86 (m, 1H), 3.77-3.73 (m, 1H), 2.42-2.32 (m, 1H), 2.33 (d, 2H), 2.05-1.94 (m, 2H), 1.86 (m, 1H), 1.76-1.71 (m, 1H), 1.60-1.54 (m, 2H), 1.02 (d, 6H); LCMS: 283 (M+H)+.
  • Example 7 Preparation of 3-(1-(Tetrahydro-2H-pyran-2-yl)-1H-indazol-5-yl)prop-2-yn-1-ol (Intermediate 16)
  • Figure US20120071535A1-20120322-C00175
  • A 500 mL three-necked round bottom flask was charged with 5-bromo-1-(tetrahydro-2H-pyran-2-yl)-1H-indazole (14.0 g, 50 mmol; Intermediate 1) and triethylamine (300 mL). The flask was degassed with 3 cycles of vacuum/N2, followed by the addition of Pd(PPh3)2Cl2 (3.5 g, 5 mmol) and CuI (0.95 g, 5 mmol) under N2 atmosphere. The flask was again degassed with 3 cycles of vacuum/N2. Prop-2-yn-1-ol (8.4 g, 150 mmol) was added via syringe and the reaction mixture was stirred at 80° C. for 16 hours. Upon completion, the solvent was evaporated. The residue was diluted with dichloromethane (400 mL), washed with water (3×200 mL) and brine (200 mL), dried over anhydrous sodium sulfate, and concentrated in vacuo. The residue was further purified on silica gel column (1:100-1:20 EtOAc/petroleum ether) affording the title compound (11.1 g). 1H NMR (400 MHz, DMSO-d6): δ 8.13 (s, 1H), 7.90 (s, 1H), 7.75 (d, 1H), 7.44 (d, 1H), 5.86 (dd, 1H), 5.33 (t, 1H), 4.33 (d, 2H), 3.89-3.86 (m, 1H), 3.79-3.73 (m, 1H), 2.45-2.35 (m, 1H), 2.05-1.95 (m, 2H), 1.80-1.70 (m, 1H), 1.60-1.56 (m, 2H); LCMS: 257 (M+H)+.
  • Example 8 Preparation of: 4-(1-(Tetrahydro-2H-pyran-2-yl)-1H-indazol-5-yl)but-3-yn-1-ol (Intermediate 17)
  • Figure US20120071535A1-20120322-C00176
  • To a mixture of 5-bromo-1-(tetrahydro-2H-pyran-2-yl)-1H-indazole (17.0 g, 60.7 mmol; Intermediate 1), Pd(PPh3)2Cl2 (5.80 g, 6.07 mmol), CuI (1.20 g, 6.07 mmol), and triethylamine (170 mL) was added but-3-yn-1-ol (6.80 g, 97.2 mmol) under N2 atmosphere. The resulting mixture was stirred at 80° C. for 16 hours under N2 atmosphere. Upon completion, the reaction mixture was diluted with EtOAc and washed with water (3×50 mL). The organic layer was dried over Na2SO4 and concentrated in vacuo. The residue was purified by column chromatography on silica gel (30-50% EtOAc in petroleum ether) affording the title compound (8.0 g). 1H NMR (400 MHz, DMSO-d6): δ 8.09 (s, 1H), 7.83 (s, 1H), 7.70 (d, 1H), 7.40 (d, 1H), 5.84 (dd, 1H), 4.90 (br, 1H), 3.91-3.87 (m, 1H), 3.77-3.70 (m, 1H), 3.60 (t, 2H), 2.56 (t, 2H), 2.48-2.33 (m, 1H), 2.04-1.94 (m, 2H), 1.76-1.69 (m, 1H), 1.60-1.55 (m, 2H); LCMS: 271 (M+H)+.
  • Example 9 Preparation of 5-Bromo-4-fluoro-1-(tetrahydro-2H-pyran-2-yl)-1H-indazole (Intermediate 18) Step 1: 4-Bromo-3-fluoro-2-methylaniline
  • Figure US20120071535A1-20120322-C00177
  • To a solution of 3-fluoro-2-methylaniline (20 g, 0.16 mol) in CH3CN (500 mL) was added NBS (31.3 g, 0.176 mol) in portions at 10° C. The resulting mixture was stirred at room temperature for 30 minutes. Upon completion, saturated Na2S2O3 (500 mL) was added slowly into the reaction mixture at 10° C. The organic layer was separated, and the aqueous layer was extracted with EtOAc. The combined organic layers were dried over Na2SO4 and concentrated in vacuo. The residue was washed with petroleum ether affording the title compound (20 g), which was used in the next step without further purification. 1H NMR (300 MHz, DMSO-d6): δ 7.08 (t, 1H), 6.40 (dd, 1H), 5.35 (br, 2H), 1.98 (d, 3H).
  • Step 2: 5-Bromo-4-fluoro-1H-indazole
  • Figure US20120071535A1-20120322-C00178
  • To a solution of 4-bromo-3-fluoro-2-methylaniline (20 g, 98.0 mmol) in CH3CO2H (600 mL) was added NaNO2 (8.1 g, 118 mmol) at 10° C. The resulting mixture was stirred at room temperature for 4 hours. Upon completion, aqueous NaOH (50%) was added to the reaction mixture until pH was ˜7-8. The mixture was extracted with EtOAc. The organic layer was dried over Na2SO4 and concentrated in vacuo. The residue was purified by column chromatography on silica gel (0˜40% EtOAc in petroleum ether) affording the title compound (16 g). 1H NMR (300 MHz, DMSO-d6): δ 13.58 (br, 1H), 8.22 (s, 1H), 7.53 (t, 1H), 7.38 (d, 1H).
  • Step 3: 5-Bromo-4-fluoro-1-(tetrahydro-2H-pyran-2-yl)-1H-indazole
  • Figure US20120071535A1-20120322-C00179
  • To a mixture of 5-bromo-4-fluoro-1H-indazole (50 g, 0.23 mol) and DHP (23 g, 0.28 mol) in dry dichloromethane (1000 mL) was added PTSA (2.2 g, 11.5 mmol) at room temperature. The resulting mixture was stirred overnight at that temperature. Upon completion, saturated aqueous NaHCO3 (100 mL) was added slowly into the reaction mixture. The organic layer was separated, dried over Na2SO4, and concentrated in vacuo. The residue was purified by column chromatography on silica gel (0-2% EtOAc in petroleum ether) and then re-crystallized from petroleum ether to afford the title compound (55 g). 1H NMR (300 MHz, DMSO-d6,): δ 8.28 (s, 1H), 7.58-7.66 (m, 2H), 5.89 (dd, 1H), 3.90-3.85 (m, 1H), 3.79-3.70 (m, 1H), 2.42-2.29 (m, 1H), 2.06-1.94 (m, 2H), 1.77-1.68 (m, 1H), 1.60-1.53 (m, 2H); LCMS: 299 (M+H)+.
  • Example 10 Preparation of 5-(Cyclopropylethynyl)-4-fluoro-1-(tetrahydro-2H-pyran-2-yl)-1H-indazole (Intermediate 19)
  • Figure US20120071535A1-20120322-C00180
  • In a high pressure tube, a mixture of 5-bromo-4-fluoro-1-(tetrahydro-2H-pyran-2-yl)-1H-indazole (8.0 g, 26.8 mmol; Intermediate 18), PdCl2(PPh3)2 (3.7 g, 5.35 mmol), CuI (1.0 g, 5.35 mmol), and triethylamine (30 mL) was deoxygenated with three cycles of vacuum/nitrogen. Ethynylcyclopropane (8.9 g, 134 mmol) was added under N2 atmosphere. The tube was sealed and the reaction mixture was heated at 120° C. for 63 hours. Upon completion, the reaction mixture was diluted with ethyl acetate and filtered through Celite. The filtrate was concentrated in vacuo and the residue was purified by column chromatography on silica gel (0-10% ethyl acetate in petroleum ether) affording the title compound (4.3 g). 1H NMR (400 MHz, DMSO-d6): δ 8.25 (s, 1H), 7.55 (d, 1H), 7.40 (dd, 1H), 5.88 (dd, 1H), 3.88-3.85 (m, 1H), 3.76-3.73 (m, 2H), 2.43-2.33 (m, 1H), 2.05-1.95 (m, 2H), 1.76-1.72 (m, 1H), 1.62-1.56 (m, 3H), 0.93-0.89 (m, 2H), 0.79-0.74 (m, 2H); LCMS: 285 (M+H)+.
  • The Intermediates in Table 4 were prepared from Intermediate 1 following the procedures outlined for Intermediates 14-17, and 19.
  • TABLE 4
    Intermediate 20 5-(Cyclopentylethynyl)-1-(tetrahydro-2H- pyran-2-yl)-1H-indazole
    Figure US20120071535A1-20120322-C00181
    Intermediate 21 5-(Cyclohexylethynyl)-1-(tetrahydro-2H- pyran-2-yl)-1H-indazole
    Figure US20120071535A1-20120322-C00182
    Intermediate 22 5-(3-Methylbut-1-yn-1-yl)-1-(tetrahydro-2H- pyran-2-yl)-1H-indazole
    Figure US20120071535A1-20120322-C00183
    Intermediate 23 5-(Hex-1-yn-1-yl)-1-(tetrahydro-2H-pyran-2- yl)-1H-indazole
    Figure US20120071535A1-20120322-C00184
    Intermediate 24 5-(3-Cyclopentylprop-1-yn-1-yl)-1- (tetrahydro-2H-pyran-2-yl)-1H-indazole
    Figure US20120071535A1-20120322-C00185
  • Example 11 Preparation of 5-(4-Chlorobut-1-yn-1-yl)-1-(tetrahydro-2H-pyran-2-yl)-1H-indazole (Intermediate 25)
  • Figure US20120071535A1-20120322-C00186
  • To a solution of 4-(1-(tetrahydro-2H-pyran-2-yl)-1H-indazol-5-yl)but-3-yn-1-ol (1.0 g, 3.7 mmol; Intermediate 17) in dry pyridine (10 mL) was added dropwise POCl3 (2.4 g, 14.7 mmol) under N2 atmosphere. The resulting solution was stirred at room temperature for 16 hours. Upon completion, the reaction mixture was concentrated in vacuo. The residue was poured into ice-water and extracted with EtOAc (2×10 mL). The combined organic layers were washed with brine, dried over Na2SO4 and concentrated in vacuo. The residue was further purified on silica gel column (0-20% EtOAc in petroleum ether) affording the title compound (400 mg). 1H NMR (400 MHz, DMSO-d6): δ 8.11 (s, 1H), 7.86 (s, 1H), 7.72 (d, 1H), 7.42 (dd, 1H), 5.86 (dd, 1H), 3.90-3.86 (m, 1H), 3.81 (t, 2H), 3.77-3.70 (m, 1H), 2.93 (t, 2H), 2.41-2.34 (m, 1H), 2.05-1.94 (m, 2H), 1.75-1.71 (m, 1H), 1.60-1.55 (m, 2H); LCMS: 289 (M+H)+.
  • Example 12 Preparation of 5-(3,3-Difluoroprop-1-yn-1-yl)-1-(tetrahydro-2,1-pyran-2-yl)-1H-indazole (Intermediate 26) Step 1: 3-(1-(Tetrahydro-2H-pyran-2-yl)-1H-indazol-5-yl)propiolaldehyde
  • Figure US20120071535A1-20120322-C00187
  • A 500 mL three-necked round bottom flask was charged with 3-(1-(tetrahydro-2H-pyran-2-yl)-1H-indazol-5-yl)prop-2-yn-1-ol (11.4 g, 44.2 mmol; Intermediate 16), dichloromethane (300 mL) and MnO2 (38.4 g, 442 mmol). The resulting mixture was stirred at room temperature for 16 hours. Upon completion, the reaction mixture was filtered. The filtrate was dried over anhydrous sodium sulfate and concentrated in vacuo. The residue was purified by column chromatography on silica gel (1:100-1:20 EtOAc/petroleum ether) affording the title compound (6.4 g). 1H NMR (400 MHz, DMSO-d6): δ 9.45 (s, 1H), 8.27-8.25 (m, 2H), 7.87 (d, 1H), 7.66 (dd, 1H), 5.86 (dd, 1H), 3.91-3.87 (m, 1H), 3.79-3.72 (m, 1H), 2.40-2.36 (m, 1H), 2.05-1.96 (m, 2H), 1.78-1.72 (m, 1H), 1.61-1.56 (m, 2H); LCMS: 255 (M+H)+.
  • Step 2: 5-(3,3-Difluoroprop-1-yn-1-yl)-1-(tetrahydro-2H-pyran-2-yl)-1H-indazole
  • Figure US20120071535A1-20120322-C00188
  • A 500 mL round bottom flask was charged with dry dichloromethane (200 mL), triethylamine.3HF (8.06 g, 50.1 mmol) and XtalFluor-E (8.61 g, 37.6 mmol) under N2 atmosphere. The resulting solution was stirred at room temperature for 10 minutes. 3-(1-(Tetrahydro-2H-pyran-2-yl)-1H-indazol-5-yl)propiolaldehyde (3.21 g, 12.5 mmol) was added, and the mixture was stirred at room temperature for 1 hour. Upon completion, saturated NaHCO3 (100 mL) was added into the mixture. The organic layer was separated, and the aqueous layer was extracted with dichloromethane (2×100 mL). The combined organic layers were dried over Na2SO4 and concentrated in vacuo. The residue was purified by column chromatography on silica gel (1:100-1:20 EtOAc/petroleum ether) affording the title compound (1.71 g). 1H NMR (400 MHz, DMSO-d6): δ 8.20 (s, 1H), 8.14 (s, 1H), 7.83 (d, 1H), 7.58 (dd, 1H), 6.99 (t, 1H), 5.90 (dd, 1H), 3.90-3.87 (m, 1H), 3.79-3.73 (m, 1H), 2.45-2.37 (m, 1H), 2.05-1.96 (m, 2H), 1.79-1.65 (m, 1H), 1.60-1.56 (m, 2H); LCMS: 277 (M+H)+.
  • Example 13 Preparation of 5-(4-Fluorobut-1-yn-1-yl)-1-(tetrahydro-2H-pyran-2-yl)-1H-indazole (Intermediate 27)
  • Figure US20120071535A1-20120322-C00189
  • To a solution of 4-(1-(tetrahydro-2H-pyran-2-yl)-1H-indazol-5-yl)but-3-yn-1-ol (1.0 g, 3.7 mmol; Intermediate 17) in dry dichloromethane (25 mL), was added triethylamine.3HF (1.2 g, 7.4 mmol). XtalFluor-E (1.2 g, 5.5 mmol) was then added. The resulting solution was stirred at room temperature for 30 minutes. Upon completion, the reaction solution was neutralized by slow addition of saturated NaHCO3 (10 mL). The organic layer was dried over Na2SO4 and concentrated in vacuo. The residue was purified by column chromatography on silica gel (0-20% EtOAc in petroleum ether) affording the title compound (100 mg). 1H NMR (400 MHz, DMSO-d6): δ 8.12 (s, 1H), 7.88 (s, 1H), 7.73 (d, 1H), 7.42 (dd, 1H), 5.86 (dd, 1H), 4.60 (dt, 2H), 3.88 (m, 1H), 3.78-3.71 (m, 1H), 2.89 (dt, 2H), 2.48-2.34 (m, 1H), 2.06-1.95 (m, 2H), 1.78-1.72 (m, 1H), 1.58 (m, 2H); LCMS: 273 (M+H)+.
  • Example 14 Preparation of 1-(Tetrahydro-2H-pyran-2-yl)-5-(3,3,3-trifluoroprop-1-yn-1-yl)-1H-indazole (Intermediate 28) Step 1: 5-Iodo-1H-indazole
  • Figure US20120071535A1-20120322-C00190
  • To a solution of 4-iodo-2-methylaniline (1.09 g, 4.68 mmol) in CH3CO2H (40 mL), were added NaNO2 (0.39 g, 5.65 mmol) and water (1 mL) at 10° C. The resulting mixture was stirred at room temperature for 6 hours. Upon completion, the reaction mixture was extracted with EtOAc. The combined organic layers were dried over Na2SO4 and concentrated in vacuo. The residue was purified by column chromatography on silica gel (0-40% EtOAc in petroleum ether) affording the title compound (0.90 g). 1H NMR (DMSO-d6, 400 MHz): δ 13.23 (br, 1H), 8.18 (s, 1H), 8.02 (s, 1H), 7.57 (d, 1H), 7.41 (d, 1H).
  • Step 2: 5-Iodo-1-(tetrahydro-2H-pyran-2-yl)-1H-indazole
  • Figure US20120071535A1-20120322-C00191
  • To a mixture of 5-iodo-1H-indazole (0.90 g, 3.69 mmol) and DHP (1.57 g, 18.7 mmol) in dry dichloromethane (20 mL), was added PTSA (0.08 g, 0.41 mmol) at room temperature. The resulting mixture was stirred overnight. Upon completion, saturated aqueous NaHCO3 (30 mL) was added slowly into the reaction mixture. The organic layer was separated, dried over Na2SO4, and concentrated in vacuo. The residue was purified by column chromatography on silica gel (0-5% EtOAc in petroleum ether) affording the title compound (1.0 g). 1H NMR (400 MHz, DMSO-d6): δ 8.21 (s, 1H), 8.08 (s, 1H), 7.67 (dd, 1H), 7.61 (d, 1H), 5.85 (dd, 1H), 3.88-3.85 (m, 1H), 3.78-3.72 (m, 1H), 2.41-2.29 (m, 1H), 2.05-1.95 (m, 2H), 1.77-1.72 (m, 1H), 1.61-1.56 (m, 2H).
  • Step 3: 1-(Tetrahydro-2H-pyran-2-yl)-5-(3,3,3-trifluoroprop-1-yn-1-yl)-1H-indazole
  • Figure US20120071535A1-20120322-C00192
  • To a solution of LDA (2 M in THF, 3.2 mL, 6.4 mmol) in anhydrous THF (10 mL) was added dropwise 2-bromo-3,3,3-trifluoroprop-1-ene (0.55 g, 3.1 mmol) at −78° C. The resulting mixture was stirred at that temperature for 15 minutes, followed by the addition of ZnCl2 (1 M in ethyl ether, 6.5 mL, 6.5 mmol) and TMEDA (1 mL, 6.5 mmol). The mixture was stirred at −78° C. for further 30 minutes and then 30 minutes at room temperature. 5-Iodo-1-(tetrahydro-2H-pyran-2-yl)-1H-indazole (0.99 g, 3.0 mmol) and Pd(PPh3)4 (0.21 g, 0.18 mmol) were added. The reaction mixture was heated at 80° C. for 6 hours under N2 atmosphere. Upon completion, the reaction mixture was quenched with water (100 mL) and then diluted with ethyl acetate (300 mL). The organic layer was separated, dried over Na2SO4 and concentrated in vacuo. The residue was purified by column chromatography on silica gel (0-=20% EtOAc in petroleum ether) affording the title compound (299 mg). 1H NMR (300 MHz, DMSO-d6): δ 8.28 (s, 1H), 8.24 (s, 1H), 7.87 (d, 1H), 7.68 (dd, 1H), 5.91 (dd, 1H), 3.90-3.86 (m, 1H), 3.79-3.72 (m, 1H), 2.41-2.36 (m, 1H), 2.05-1.96 (m, 2H), 1.76-1.72 (m, 1H), 1.60-1.56 (m, 2H); LCMS: 295 (M+H)+.
  • Example 15 Preparation of 5-(4-Chlorobut-1-yn-1-yl)-4-fluoro-1-(tetrahydro-2H-pyran-2-yl)-1H-indazole (Intermediate 29) Step 1: 4-(4-Fluoro-1-(tetrahydro-2H-pyran-2-yl)-1H-indazol-5-yl)but-3-yn-1-ol
  • Figure US20120071535A1-20120322-C00193
  • To a mixture of 5-bromo-4-fluoro-1-(tetrahydro-2H-pyran-2-yl)-1H-indazole (2.80 g, 9.36 mmol, Intermediate 18), Pd(PPh3)2Cl2 (660 mg, 0.94 mmol), CuI (180 mg, 0.94 mmol), and triethylamine (50 mL) was added but-3-yn-1-ol (2.0 g, 28.1 mmol) under N2 atmosphere. The resulting mixture was stirred at 60° C. for 16 hours. Upon completion, the reaction mixture was diluted with EtOAc, and washed with water (3×10 mL). The organic layer was dried over Na2SO4 and concentrated in vacuo. The residue was purified on silica gel column (0-20% EtOAc in petroleum ether) affording the title compound (2.0 g). 1H NMR (300 MHz, DMSO-d6): δ 8.33 (s, 1H), 7.64 (d, 1H), 7.49 (dd, 1H), 5.94 (dd, 1H), 4.99 (t, 1H), 3.98-3.92 (m, 1H), 3.85-3.77 (m, 1H), 3.68 (t, 2H), 2.67 (t, 2H), 2.48-2.35 (m, 1H), 2.12-2.02 (m, 2H), 1.84-1.78 (m, 1H), 1.68-1.62 (m, 2H).
  • Step 2: 5-(4-Chlorobut-1-yn-1-yl)-4-fluoro-1-(tetrahydro-2H-pyran-2-yl)-1H-indazole
  • Figure US20120071535A1-20120322-C00194
  • To a mixture of 4-(4-fluoro-1-(tetrahydro-2H-pyran-2-yl)-1H-indazol-5-yl)but-3-yn-1-ol (4.6 g, 16.0 mmol) in pyridine (50 mL), was added POCl3 (10.3 g, 67.3 mmol). The resulting solution was stirred at room temperature for 2 hours. Upon completion, the reaction solution was poured into water (250 mL) and extracted with EtOAc. The organic layer was dried over Na2SO4 and concentrated in vacuo. The residue was purified on silica gel column (0-10% EtOAc in petroleum ether) affording the title compound (2.62 g). 1H NMR (300 MHz, DMSO-d6): δ 8.13 (s, 1H), 7.58 (d, 1H), 7.43 (dd, 1H), 5.87 (dd, 1H), 3.90-3.69 (m, 2H), 3.83 (t, 2H), 2.98 (t, 1H), 2.41-2.29 (m, 1H), 2.06-1.94 (m, 2H), 1.78-1.70 (m, 1H), 1.60-1.54 (m, 2H); LCMS: 307 (M+H)+.
  • Example 16 Preparation of 4-Fluoro-5-(4-fluorobut-1-yn-1-yl)-1-(tetrahydro-2H-pyran-2-yl)-1H-indazole (Intermediate 30)
  • Figure US20120071535A1-20120322-C00195
  • To a solution of 4-(4-fluoro-1-(tetrahydro-2H-pyran-2-yl)-1H-indazol-5-yl)but-3-yn-1-ol (3.4 g, 11.8 mmol; Intermediate 29, Step 1) in dry dichloromethane (100 mL), was added triethylamine.3HF (7.6 g, 47.2 mmol). XtalFluor-E (8.0 g, 34.9 mmol) was then added. The resulting solution was stirred at room temperature for 30 minutes. Upon completion, the reaction solution was neutralized by slow addition of saturated NaHCO3 (30 mL). The organic layer was dried over Na2SO4 and concentrated in vacuo. The residue was purified by column chromatography on silica gel (0-20% EtOAc in petroleum ether), and then re-crystallized from petroleum ether to afford the title compound (1.3 g). 1H NMR (300 MHz, DMSO-d6): δ 8.27 (s, 1H), 7.58 (d, 1H), 7.44 (dd, 1H), 5.89 (dd, 1H), 4.60 (dt, 2H), 3.91-3.85 (m, 1H), 3.80-3.69 (m, 1H), 2.93 (dt, 2H), 2.46-2.28 (m, 1H), 2.06-1.95 (m, 2H), 1.78-1.67 (m, 1H), 1.60-1.52 (m, 2H); LCMS: 291 (M+H)+.
  • Example 17 Preparation of: 5-(3-Methoxyprop-1-yn-1-yl)-1-(tetrahydro-2H-pyran-2-yl)-1H-indazole (Intermediate 31)
  • Figure US20120071535A1-20120322-C00196
  • Sodium hydride (60% in mineral oil, 0.42 g, 10.5 mmol) was added to a solution of 3-(1-(tetrahydro-2H-pyran-2-yl)-1H-indazol-5-yl)prop-2-yn-1-ol (1.01 g, 3.94 mmol; Intermediate 16) in THF (20 mL) at 0° C. The mixture was stirred at 0° C. for 30 minutes, and then iodomethane (1.67 g, 11.8 mmol) was added. The resulting mixture was stirred at room temperature for 16 h. The reaction mixture was poured into ice water (100 mL) and extracted with EtOAc (3×100 mL). The combined organic layers were washed with brine (100 mL), dried over anhydrous sodium sulfate, and concentrated in vacuo. The residue was purified by column chromatography on silica gel (EtOAc/petroleum ether=1:30) to afford the title compound (0.714 g, yield 67.3%). 1H NMR (DMSO-d6, 300 MHz): δ 8.21 (s, 1H), 7.93 (s, 7.73 (d, 1H), 7.47-7.45 (m, 1H), 5.85-5.83 (m, 1H), 4.38 (s, 2H), 3.76-3.71 (m, 2H), 3.35 (s, 3H), 2.40-2.37 (m, 1H), 2.03-1.94 (m, 2H), 1.73-1.72 (m, 1H), 1.57-1.55 (m, 2H). LCMS: 271 (M+H)+.
  • The Intermediates in Table 5 were prepared from Intermediate 1 following the procedures outlined for Intermediates 16, 17 & 31.
  • TABLE 5
    Intermediate 32 5-(4-Methoxybut-1-yn-1-yl)-1-(tetrahydro- 2H-pyran-2-yl)-1H-indazole
    Figure US20120071535A1-20120322-C00197
    Intermediate 33 5-(5-Methoxypent-1-yn-1-yl)-1- (tetrahydro-2H-pyran-2-yl)-1H-indazole
    Figure US20120071535A1-20120322-C00198
    Intermediate 34 5-(6-Methoxyhex-1-yn-1-yl)-1- (tetrahydro-2H-pyran-2-yl)-1H-indazole
    Figure US20120071535A1-20120322-C00199
  • Example 18 Preparation of But-1-yn-1-yltrimethylsilane (Intermediate 35)
  • Figure US20120071535A1-20120322-C00200
  • A 3 L three-necked round bottom flask was charged with (trimethylsilyl)acetylene (116 g, 1.19 mol) and dry THF (400 mL). The solution was cooled to −78° C. To this solution, butyllithium in hexane (2.5 M, 500 mL, 1.25 mol) was added dropwise over 2 hours. The resulting mixture was warmed to 0° C. for 10 minutes and then re-cooled to −78° C. HMPA (234 g, 1.31 mol) was added, and the mixture was stirred at −78° C. for 30 minutes. To this solution, iodoethane (200 g, 1.28 mol) was added. The reaction mixture was allowed to warm to room temperature and stirred overnight. Upon completion, the reaction mixture was washed with water (4×600 mL) and then brine (2×500 mL). The organic layer was dried over anhydrous sodium sulfate and filtered. Hexane and THF were distilled off at 75110° C. But-1-yn-1-yltrimethylsilane was distilled between 125 to 135° C. affording 91 g of a colorless liquid (61%). 1H NMR (400 MHz, DMSO-d6) δ 2.20 (q, 2H), 1.05 (t, 3H), 0.11 (s, 9H); 13C NMR (100 MHz, CDCl3): δ 108.8, 83.3, 13.7, 13.4, 0.0.
  • Example 19 Alternate Preparation of Intermediate 3
  • Figure US20120071535A1-20120322-C00201
  • A mixture of 5-bromo-1-(tetrahydro-2H-pyran-2-yl)-1H-indazole (39.6 g, 0.142 mol; Intermediate 1), Cs2CO3 (60.0 g, 184 mmol), CuI (1.35 g, 7.08 mmol), Pd(OAc)2 (1.59 g, 7.08 mmol), dppf (3.93 g, 7.08 mmol), and DMA (160 mL) was degassed with three vacuum/nitrogen cycles. But-1-yn-1-yltrimethylsilane (23.2 g, 184 mmol; Intermediate 35) was added, and the resulting mixture was heated at 80° C. for 5 h under N2. Upon completion by LCMS, the reaction mixture was diluted with EtOAc (300 mL) and H2O (300 mL) and then filtered. The organic layer of the filtrate was separated, and the aqueous layer was extracted with EtOAc (3×150 mL). The combined organic layers were washed with brine (2×100 mL), dried over anhydrous Na2SO4, filtered and concentrated. The residue was purified on silica gel column (300-400 mesh, 20 cm in diameter and 15 cm in height) using EtOAc/petroleum ether (1 L of petroleum ether; then 1 L of EtOAc/petroleum ether=1/50; and then EtOAc/Petroleum ether=1/30 until the by-product was washed out; then EtOAc/petroleum ether=1/10 to collect the product) affording a yellow oil (33 g) which solidified over time in the 4° C. refrigerator. The resulting solid was further washed with petroleum ether (200 mL, then 3×50 mL) affording the title compound as an off-white solid (26 g, 73%).
  • Example 20 Preparation of 5-(But-1-yn-1-yl)-4-fluoro-1-(tetrahydro-2,1-pyran-2-yl)-1H-indazole (Intermediate 36)
  • Figure US20120071535A1-20120322-C00202
  • Nitrogen was bubbled into a solution of 5-bromo-4-fluoro-1-(tetrahydro-2H-pyran-2-yl)-1H-indazole (19.7 g, 65.9 mmol; Intermediate 18) and DMA (60 mL). After 5 min, CuI (1.25 g, 6.6 mmol), Pd(OAc)2 (1.48 g, 6.6 mmol), dppf (3.66 g, 6.6 mmol), Cs2CO3 (34.3 g, 105.4 mmol), and but-1-yn-1-yltrimethylsilane (11.6 g, 92.3 mmol; Intermediate 35) were added sequentially with continued N2 bubbling. The resulting mixture was heated at 80° C. for 18 h under N2. The reaction mixture was diluted with EtOAc (900 mL) and H2O (500 mL) and then filtered. The organic layer of the filtrate was separated, and the aqueous layer was extracted with EtOAc (2×100 mL). The combined organic layers were washed with brine (3×100 mL), dried over anhydrous Na2SO4, concentrated, and purified by silica gel chromatography (1:30 EtOAc/petroleum ether) to give the title compound (15.2 g) as a white solid. 1H NMR (DMSO-d6, 400 MHz): δ 8.26 (s, 1H), 7.57 (d, 1H), 7.42 (dd, 1H), 5.87 (dd, 1H), 3.90-3.86 (m, 1H), 3.78-3.71 (m, 1H), 2.48 (q, 2H), 2.40-2.30 (m, 1H), 2.05-1.95 (m, 2H), 1.77-1.71 (m, 1H), 1.59-1.57 (m, 2H), 1.19 (t, 3H); LCMS: 273 (M+H)+.
  • Example 21 Preparation of 5-(But-1-yn-1-yl)-4-chloro-1-(tetrahydro-2H-pyran-2-yl)-1H-indazole (Intermediate 37) Step 1: 4-Bromo-3-chloro-2-methylaniline
  • Figure US20120071535A1-20120322-C00203
  • To a solution of 3-chloro-2-methylaniline (30 g, 0.212 mol) in CH3CN (300 mL) was added NBS (45.2 g, 0.254 mol) in portions at 10° C. The resulting mixture was stirred at room temperature for 30 minutes. Upon completion, saturated Na2S2O3 (500 mL) was added slowly into the reaction mixture at 10° C. The organic layer was separated, and the aqueous layer was extracted with EtOAc. The combined organic layers were dried over Na2SO4 and concentrated in vacuo. The residue was washed with petroleum ether to afford the title compound (30 g), which was used in the next step without further purification. 1H NMR (300 MHz, CDCl3): δ 7.24 (d, 1H), 6.48 (d, 1H), 3.70 (br, 2H), 2.28 (s, 3H).
  • Step 2: 5-Bromo-4-chloro-1H-indazole
  • Figure US20120071535A1-20120322-C00204
  • To a solution of 4-bromo-3-chloro-2-methylaniline (11 g, 49.9 mmol) in CH3CO2H (450 mL) was added NaNO2 (5.4 g, 78.3 mmol) in H2O (15 mL) at 10° C. The resulting mixture was stirred at room temperature for 30 minutes. Upon completion, the reaction mixture was diluted with H2O (500 mL) and extracted with EtOAc. The organic layer was dried over Na2SO4 and concentrated in vacuo. The residue was triturated with petroleum ether affording the title compound (4.5 g) as yellow solid. 1H NMR (400 MHz, DMSO-d6): δ 13.60 (s, 1H), 8.15 (s, 1H), 7.62 (d, 1H), 7.52 (d, 1H).
  • Step 3: 5-Bromo-4-chloro-1-(tetrahydro-2H-pyran-2-yl)-1H-indazole
  • Figure US20120071535A1-20120322-C00205
  • To a mixture of 5-bromo-4-chloro-1H-indazole (8.0 g, 34.6 mmol) and DHP (8.72 g, 0.104 mol) in dry dichloromethane (200 mL) was added PTSA (0.657 g, 3.46 mmol) at room temperature. The resulting mixture was stirred at room temperature overnight. Upon completion, saturated aqueous NaHCO3 (100 mL) was added slowly to the reaction mixture. The organic layer was separated, dried over Na2SO4, and concentrated in vacuo. The residue was purified by column chromatography on silica gel (0-3% EtOAc in petroleum ether) affording the title compound (8.9 g). 1H NMR (300 MHz, DMSO-d6): δ 8.19 (s, 1H), 7.71 (m, 2H), 5.88 (dd, 1H), 3.89-3.84 (m, 1H), 3.79-3.73 (m, 1H), 2.42-2.32 (m, 1H), 2.05-1.95 (m, 2H), 1.75-1.70 (m, 1H), 1.60-1.54 (m, 2H).
  • Step 4: 5-(But-1-yn-1-yl)-4-chloro-1-(tetrahydro-2,1-pyran-2-yl)-1H-indazole
  • Figure US20120071535A1-20120322-C00206
  • In a 20 mL microwave tube, nitrogen was bubbled through triethylamine (6 mL) for 10 minutes. 5-bromo-4-chloro-1-(tetrahydro-2H-pyran-2-yl)-1H-indazole (2.00 g, 6.34 mmol), tetrabutylammonium fluoride (3.70 g, 14.3 mmol), CuI (0.24 g, 1.3 mmol) and Pd(PPh3)4 (1.46 g, 1.26 mmol) were added under nitrogen atmosphere, and bubbling of nitrogen was continued for another 5 minutes. But-1-yn-1-yltrimethylsilane (1.80 g, 14.3 mmol; Intermediate 35) was then added and the tube was sealed immediately. The reaction mixture was heated in a microwave reactor at 120° C. for 3 hours. Four of these reactions (4×2 g scale per run) were combined, mixed with water (100 mL), and extracted with ethyl acetate (2×100 mL). The combined organic layers were dried over Na2SO4 and concentrated in vacuo. The residue was purified by column chromatography on silica gel (0-3% ethyl acetate in petroleum ether) affording the title compound (4.5 g, 90%), which was then triturated with petroleum ether (8 mL). The solid was collected and dried to afford a pale yellow powder (3.5 g). This powder was re-crystallized from ethyl acetate (2 mL) to afford the pure title compound as pale yellow crystals (3.0 g). 1H NMR (400 MHz, DMSO-d6): δ 8.19 (s, 1H), 7.72 (d, 1H), 7.49 (d, 1H), 5.88 (dd, 1H), 3.89-3.85 (m, 1H), 3.79-3.73 (m, 1H), 2.50 (q, 2H), 2.43-2.33 (m, 1H), 2.05-1.95 (m, 2H), 1.75-1.70 (m, 1H), 1.60-1.55 (m, 2H), 1.21 (t, 3H); LCMS: 289 (M+H)+.
  • Example 22 Preparation of 5-(But-1-yn-1-yl)-7-fluoro-1-(tetrahydro-2H-pyran-2-yl)-1H-indazole (Intermediate 38) Step 1: 5-Bromo-2,3-difluorobenzaldehyde
  • Figure US20120071535A1-20120322-C00207
  • To a solution of 2,3-difluorobenzaldehyde (42 g, 0.296 mol) in H2SO4 (150 mL), was added NBS (63 g, 0.354 mol) in three portions over a period of 30 minutes at 60° C. The resulting mixture was heated for 6 hours at this temperature under N2. Work-up: the reaction mixture was poured into ice water. Petroleum ether (300 mL) was added, and the mixture was stirred for 10 minutes. The organic layer was separated, and the aqueous layer was extracted with more petroleum ether (300 mL). The combined organic layers were dried over Na2SO4 and concentrated in vacuo. The residue was purified by column chromatography on silica gel (0-0.5% EtOAc in petroleum ether) to give 5-bromo-2,3-difluorobenzaldehyde (17.4 g). 1H NMR (300 MHz, CDCl3): δ 10.32 (s, 1H), 7.81-7.79 (m, 1H), 7.65-7.60 (m, 1H).
  • Step 2: (E)-5-Bromo-2,3-difluorobenzaldehyde O-methyl oxime
  • Figure US20120071535A1-20120322-C00208
  • A mixture of 5-bromo-2,3-difluorobenzaldehyde (17.38 g, 78.6 mmol), O-methylhydroxylamine hydrochloride (7.23 g, 86.46 mmol), and K2CO3 (13 g, 94.32 mmol) in DME (80 mL) was heated at 40° C. for 14 h. Work-up: the reaction mixture was filtered. The filtrate was concentrated in vacuo, and the residue was purified by column chromatography on silica gel (0-2% EtOAc in petroleum ether), to give (E)-5-bromo-2,3-difluorobenzaldehyde O-methyl oxime (19.65 g). 1H NMR (CDCl3, 300 MHz): δ 8.20 (s, 1H), 7.76-7.73 (m, 1H), 7.35-7.29 (m, 1H), 4.01 (s, 3H).
  • Step 3: 5-Bromo-7-fluoro-1H-indazole
  • Figure US20120071535A1-20120322-C00209
  • A mixture of (E)-5-bromo-2,3-difluorobenzaldehyde O-methyl oxime (19.65 g, 78.6 mmol), hydrazine hydrate (80 mL), and dry THF (80 mL), was heated at 90° C. for 84 h. Work-up: the organic solvent was evaporated. The resulting mixture was diluted with EtOAc (400 mL), washed with water (150 mL), dried over Na2SO4, and concentrated in vacuo. The residue was purified by column chromatography on silica gel (0=20% EtOAc in petroleum ether) to give 5-bromo-7-fluoro-1H-indazole as a white solid (9.3 g). 1H NMR (300 MHz, CDCl3): δ 13.83 (br, 1H), 8.16 (s, 1H), 7.87 (s, 1H), 7.45 (d, 1H).
  • Step 4: 5-Bromo-7-fluoro-1-(tetrahydro-2H-pyran-2-yl)-1H-indazole
  • Figure US20120071535A1-20120322-C00210
  • To a mixture of 5-bromo-7-fluoro-1H-indazole (9.3 g, 43.26 mmol) and DHP (4.36 g, 51.9 mmol) in dry dichloromethane (100 mL), was added PTSA (424 mg, 2.16 mmol) at room temperature. The resulting mixture was stirred overnight. Work-up: saturated aqueous NaHCO3 (30 mL) was slowly added to the reaction mixture. The organic layer was separated, dried over Na2SO4, and concentrated in vacuo. The residue was purified by column chromatography on silica gel (0-10% EtOAc in petroleum ether) to give 5-bromo-7-fluoro-1-(tetrahydro-2H-pyran-2-yl)-1H-indazole as a light yellow solid. (7.8 g). 1H NMR (300 MHz, CDCl3): δ 7.98 (s, 1H), 7.64 (s, 1H), 7.22 (dd, 1H), 5.84 (dd, 1H), 4.07-4.02 (m, 1H), 3.78-3.71 (m, 1H), 2.62-2.53 (m, 1H), 2.16-2.07 (m, 2H), 1.79-1.71 (m, 2H), 1.63-1.33 (m, 1H).
  • Step 5: 5-(But-1-yn-1-yl)-7-fluoro-1-(tetrahydro-2H-pyran-2-yl)-1H-indazole
  • Figure US20120071535A1-20120322-C00211
  • Nitrogen was bubbled into a solution of 5-bromo-7-fluoro-1-(tetrahydro-2H-pyran-2-yl)-1H-indazole (7.5 g, 25.33 mmol) and DMA (100 mL). After 5 min, CuI (241 mg, 1.27 mmol), Pd(OAc)2 (284 mg, 1.27 mmol), dppf (704 mg, 1.27 mmol), K2CO3 (4.89 g, 35.46 mmol), and but-1-yn-1-yltrimethylsilane (4.46 g, 35.46 mmol) were added sequentially with continued N2 bubbling. The resulting mixture was heated at 80° C. for 10 h under N2. The reaction mixture was diluted with EtOAc (250 mL) and H2O (200 mL) and filtered. The organic layer of the filtrate was separated, and the aqueous layer was extracted with EtOAc (2×100 mL). The combined organic layers were washed with brine (3×50 mL), dried over anhydrous Na2SO4, concentrated, and purified by silica gel chromatography (1:30 EtOAc/petroleum ether) to give the pure product as a yellow solid (3.6 g) and an impure product (2 g; further purified to give additional 1.47 g). 1H NMR (300 MHz, DMSO-d6): δ 8.19 (d, 1H), 7.68 (d, 1H), 7.26 (dd, 1H), 5.79 (dd, 1H), 3.92-3.87 (m, 1H), 3.69-3.60 (m, 1H), 2.47-2.34 (m, 1H), 2.43 (q, 2H), 2.07-2.02 (m, 2H), 1.76-1.69 (m, 1H), 1.57-1.50 (m, 2H), 1.17 (t, 3H); LCMS: 273 (M+H)+.
  • Example 23 Preparation of 5-Bromo-3-fluoro-1-(tetrahydro-2H-pyran-2-yl)-1H-indazole (Intermediate 39) Step 1: 5-Bromo-3-fluoro-1H-indazole
  • Figure US20120071535A1-20120322-C00212
  • A mixture of 5-bromo-1-(tetrahydro-2H-pyran-2-yl)-1H-indazole (10.0 g, 35.7 mmol; Intermediate 1), acetic acid (4 mL), Selectfluor (25.3 g, 71.4 mmol), and acetonitrile (100 mL) was refluxed under N2 for 2 h. The reaction was allowed to cool to rt, diluted with ethyl acetate (420 mL), and then washed with water (270 mL). The organic layer was dried over Na2SO4 and concentrated in vacuo. The residue was purified on a silica gel column using EtOAc in petroleum ether (1:20) to afford the title compound as yellow solids (6.0 g, yield 78.1%). 1H NMR (DMSO-d6, 400 MHz): δ 12.77 (s, 1H), 7.96 (s, 1H), 7.54 (d, 1H), 7.48 (d, 1H).
  • Step 2: 5-Bromo-3-fluoro-1-(tetrahydro-2H-pyran-2-yl)-1H-indazole
  • Figure US20120071535A1-20120322-C00213
  • A mixture of 5-bromo-3-fluoro-1H-indazole (6.0 g, 27.9 mmol), PTSA (530.7 mg, 2.79 mmol) and DHP (3.05 g, 36.3 mmol) in dichloromethane (80 mL) was stirred at room temperature for 18 h. The reaction mixture was diluted with dichloromethane (370 mL) and washed with water (230 mL). The organic layer was dried over Na2SO4 and concentrated in vacuo. The residue was purified on a silica gel column using EtOAc in petroleum ether (1:100 to 1:15) to afford the title compound as a yellow solid (6.2 g, yield 74.3%). 1H NMR (DMSO-d6, 400 MHz): δ 7.96 (s, 1H), 7.70 (d, 1H), 7.60 (d, 1H), 5.76 (dd, 1H), 3.84-3.80 (m, 1H), 3.71-3.64 (m, 1H), 2.20-2.15 (m, 1H), 1.97-1.87 (m, 2H), 1.69-1.64 (m, 1H), 1.53-1.47 (m, 2H).
  • Example 24 Preparation of 5-(But-1-yn-1-yl)-3-fluoro-1-(tetrahydro-2H-pyran-2-yl)-1H-indazole (Intermediate 40)
  • Figure US20120071535A1-20120322-C00214
  • A 100 mL round bottom flask was charged with 5-bromo-3-fluoro-1-(tetrahydro-2H-pyran-2-yl)-1H-indazole (6.2 g, 20.7 mmol; Intermediate 39), DMA (20 mL), CuI (393.3 mg, 2.07 mmol), Pd(OAc)2 (465.0 mg, 2.07 mmol), dppf (1.1 g, 2.07 mmol), Cs2CO3 (10.8 g, 33.1 mmol), and but-1-yn-1-yltrimethylsilane (3.4 g, 26.9 mmol; Intermediate 35) sequentially while N2 was bubbled through the solution. The resulting mixture was heated at 80° C. for 10 h under N2. The reaction mixture was diluted with EtOAc (350 mL) and H2O (300 mL) and filtered. The organic layer of the filtrate was separated, and the aqueous layer was extracted with EtOAc (2×50 mL). The combined organic layers were washed with brine (3×100 mL), dried over anhydrous Na2SO4, and concentrated in vacuo. The residue was purified on a silica gel column using EtOAc in petroleum ether (1:30) to afford the title compound as a yellow solid (3.9 g, yield 69.1%). 1H NMR (DMSO-d6, 400 MHz): δ 7.76-7.71 (m, 2H), 7.49 (d, 1H), 5.79 (dd, 1H), 3.88-3.85 (m, 1H), 3.74-3.71 (m, 1H), 2.44 (q, 2H), 2.24-2.21 (m, 1H), 2.01-1.91 (m, 2H), 1.70-1.65 (m, 1H), 1.57-1.54 (m, 2H), 1.18 (t, 3H); LCMS: 273 (M+H)+.
  • Example 25 Preparation of 5-(3-Cyclopropylprop-1-yn-1-yl)-1-(tetrahydro-2H-pyran-2-yl)-1H-indazole (Intermediate 41) Step 1: (3-Cyclopropylprop-1-yn-1-yl)trimethylsilane
  • Figure US20120071535A1-20120322-C00215
  • A 500 mL three-necked round bottom flask was charged with (trimethylsilyl)acetylene (15 g, 153 mmol) and dry THF (75 mL). The solution was cooled to −78° C., and a solution of n-butyllithium in hexane (2.5 M, 75 mL, 188 mmol) was added dropwise over 30 minutes. The resulting mixture was stirred at 0° C. for 10 minutes and then re-cooled to −78° C. HMPA (40 g, 223 mmol) was added, and the mixture was stirred at −78° C. for 30 minutes. (Bromomethyl)cyclopropane (20.6 g, 153 mmol) was then added. The reaction mixture was allowed to warm to room temperature and stirred overnight. Upon completion, the reaction mixture was washed with water (4×100 mL) and brine (2×100 mL) sequentially. The organic layer was dried over anhydrous sodium sulfate. Hexane and THF was distilled off at 75˜110° C. then distillation at 138˜142° C. afforded the title compound (12 g). 1H NMR (400 MHz, DMSO-d6): δ 2.27 (d, 2H), 0.91-0.84 (m, 1H), 0.43-0.34 (m, 2H), 0.19-0.14 (m, 2H), 0.11 (s, 9H).
  • Step 2: 5-(3-Cyclopropylprop-1-yn-1-yl)-1-(tetrahydro-2,1-pyran-2-yl)-1H-indazole
  • Figure US20120071535A1-20120322-C00216
  • To a mixture of 5-bromo-1-(tetrahydro-2H-pyran-2-yl)-1H-indazole (4.0 g, 14.3 mmol; Intermediate 1), Pd(PPh3)2Cl2 (1.0 g, 1.43 mmol), CuI (271 mg, 1.43 mmol), TBAF (11.2 g, 42.8 mmol), triethylamine (20 mL), and THF (20 mL), was added (3-cyclopropylprop-1-yn-1-yl)trimethylsilane (7.9 g, 42.8 mmol) under N2 atmosphere. The resulting mixture was stirred at 80° C. for 16 hours under N2 atmosphere. Upon completion, the reaction mixture was diluted with EtOAc and filtered. The filtrate was washed with water (3×10 mL), dried over Na2SO4 and concentrated in vacuo. The residue was purified by column chromatography on silica gel (0-10% EtOAc in petroleum ether) affording the title compound as yellow solid (3.4 g). 1H NMR (400 MHz, DMSO-d6): δ 8.09 (s, 1H), 7.84 (s, 1H), 7.69 (d, 1H), 7.39 (dd, 1H), 5.83 (dd, 1H), 3.89-3.86 (m, 1H), 3.77-3.70 (m, 1H), 2.49 (d, 2H), 2.43-2.34 (m, 1H), 2.05-1.94 (m, 2H), 1.79-1.71 (m, 1H), 1.60-1.55 (m, 2H), 1.06-0.96 (m, 1H), 0.52-0.46 (m, 2H), 0.30-0.25 (m, 2H); LCMS: 281 (M+H)+.
  • Example 26 Preparation of (Cyclobutylethynyl)trimethylsilane (Intermediate 42) Step 1: (6-Chlorohex-1-yn-1-yl)trimethylsilane
  • Figure US20120071535A1-20120322-C00217
  • To a solution of 6-chlorohex-1-yne (100 mL, 94.6 g, 0.82 mol) in anhydrous Et2O (500 mL) at −78° C., n-butyllithium (2.5 M in hexane, 360 mL, 0.90 mol) was added over 40 minutes. The resulting mixture was stirred for 30 minutes at −78° C. Chlorotrimethylsilane (125 mL, 1.0 mol) was then added. The mixture was allowed to warm to room temperature and stirred for 16 h. The reaction mixture was carefully quenched with saturated aqueous NH4Cl (300 mL) at room temperature and extracted with Et2O (2×200 mL). The combined organic layers were washed with brine (200 mL), dried over anhydrous Na2SO4, and concentrated in vacuo to afford the title compound (144 g, yield 93%). 1H NMR (DMSO-d6, 400 MHz): δ 3.65 (t, 2H), 2.25 (t, 2H), 1.82-1.75 (m, 2H), 1.58-1.51 (m, 2H), 0.12 (s, 9H).
  • Step 2: (Cyclobutylethynyl)trimethylsilane
  • Figure US20120071535A1-20120322-C00218
  • To a solution of diisopropylamine (153 g, 1.52 mol) in anhydrous THF (1.0 L) at 0° C., n-butyllithium (2.5 M in hexane, 608 mL, 1.52 mol) was added dropwise. The mixture was stirred for 20 minutes at 0° C. and then cooled to −78° C. To this mixture, a solution of (6-chlorohex-1-yn-1-yl)trimethylsilane (144 g, 0.76 mol) in anhydrous THF (200 mL) was added dropwise. The resulting mixture was allowed to warm to room temperature and stirred for 16 h. The reaction mixture was carefully quenched at room temperature with saturated aqueous NH4Cl (500 mL), and then extracted with pentane (2×200 mL). The combined organic layers were washed with brine (500 mL) and dried over anhydrous Na2SO4. The solvent was evaporated on a rotary evaporator. The residue was distilled at 160-162° C./760 Torr to afford the title compound as a colorless liquid (81 g, yield 70%). 1H NMR (CDCl3, 400 MHz): δ 3.05-3.01 (m, 1H), 2.26-2.20 (m, 2H), 2.17-2.10 (m, 2H), 1.93-1.84 (m, 2H), 0.11 (s, 9H).
  • Example 27 Preparation of 5-(Cyclobutylethynyl)-1-(tetrahydro-2H-pyran-2-yl)-1H-indazole (Intermediate 43)
  • Figure US20120071535A1-20120322-C00219
  • A 100 mL round bottom flask was charged with 5-bromo-1-(tetrahydro-2H-pyran-2-yl)-1H-indazole (6.8 g, 24.2 mmol; Intermediate 1), DMA (30 mL), CuI (0.46 g, 2.4 mmol), Pd(OAc)2 (0.55 g, 2.4 mmol), dppf (1.35 g, 2.4 mmol), Cs2CO3 (11.2 g, 34.4 mmol), and (cyclobutylethynyl)trimethylsilane (5.2 g, 34.1 mmol; Intermediate 42) sequentially while N2 was bubbled through the mixture. The resulting mixture was heated at 80° C. under N2 atmosphere for 2 hours. The reaction mixture was cooled to room temperature, diluted with EtOAc (100 mL) and H2O (100 mL) and filtered. The organic layer of the filtrate was separated, and the aqueous layer was extracted with additional EtOAc (2×50 mL). The combined organic layers were washed with brine (3×100 mL), dried over anhydrous Na2SO4, and concentrated in vacuo. The residue was purified on a silica gel column using 0-10% EtOAc in petroleum ether to afford the title compound as yellow oil (4.8 g, 71%). 1H NMR (DMSO-d6): δ 8.08 (s, 1H), 7.82 (s, 1H), 7.68 (d, 1H), 7.39 (dd, 1H), 5.82 (dd, 1H), 3.89-3.85 (m, 1H), 3.76-3.69 (m, 1H), 3.30-3.24 (m, 1H), 2.39-2.26 (m, 3H), 2.19-2.09 (m, 2H), 2.03-1.84 (m, 4H), 1.75-1.70 (m, 1H), 1.58-1.55 (m, 2H); LCMS: 281 (M+H)+.
  • The Intermediate in Table 6 was prepared from Intermediate 39 & 42 following the procedure outlined for Intermediate 43.
  • TABLE 6
    Intermediate 44 5-(Cyclobutylethynyl)-3-fluoro-1- (tetrahydro-2H-pyran-2-yl)-1H-indazole
    Figure US20120071535A1-20120322-C00220

    General Procedure A: Installation of the tetrahydropyran (THP) protecting group for the indazole NH.
  • Figure US20120071535A1-20120322-C00221
  • 3,4-Dihydro-2H-pyran (1.1-10 equiv) was added to a solution of the appropriate halo-indazole (1.0 equiv), PPTS (or pTsOH, 0.05-0.3 equiv), and dichloromethane (˜2 mL/mmol) at room temperature. The reaction was stirred under N2 for 6-48 h (until complete by TLC or LCMS), quenched with water, and then extracted with dichloromethane. The extracts were dried, filtered, concentrated, and purified by silica gel chromatography to give the protected halo-indazole.
  • General Procedure B: Coupling of the protected halo-indazoles with alkynyl-trimethylsilanes
  • Figure US20120071535A1-20120322-C00222
  • A mixture of the appropriate protected halo-indazole (1.0 equiv), Cs2CO3 (1.3-3.0 equiv), CuI (0.05-0.2 equiv), Pd(OAc)2 (0.05-0.2 equiv), dppf (0.05-0.2 equiv), and N,N-dimethylacetamide (DMA, 1-2 mL/mmol) was degassed with three vacuum/nitrogen cycles. The appropriate alkynyl-trimethylsilane (1.3-2.0 equiv) was added, and the reaction was heated at 80° C. under N2 for 2-24 hours (until complete by TLC or LCMS). The reaction was allowed to cool to room temperature, diluted with ethyl acetate and water, and then filtered through Celite. The aqueous layer was separated and extracted with ethyl acetate. The organics were combined, dried, filtered, concentrated, and then purified by silica gel column chromatography to give the alkynyl-indazole.
  • Note: Alternate bases include K2CO3 and CsF; Alternate ligands include 1,3-Bis(2,4,6-trimethylphenyl)imidazolium chloride and Ph3P; Alternate catalysts include Pd(PPh3)4 and PdCl2(PPh3)2; Alternate solvents include THF and pyrrolidine. Water content seems to impact the rate of this reaction: when anhydrous Cs2CO3 and anhydrous solvent were used, 1% water (v/v with respect to solvent) was added to the reaction, and when the Cs2CO3 and/or solvent were not anhydrous, no water was added.
  • An alternate procedure has also been employed, especially with iodo-indazoles: The appropriate alkynyl-trimethylsilane (2.1 equiv) was added to a degassed solution of TBAF (2.0 equiv, 0.5M in THF). After 5-30 min, the appropriate halo-indazole (1.0 equiv), CuI (0.05-0.3 equiv), and Pd(PPh3)4 (0.05-0.2 equiv) were added. The reaction was stirred at room temperature under N2 for 2-24 hours (until complete by TLC or LCMS), then diluted with water, and extracted with an appropriate solvent. The extracts were combined, dried, filtered, concentrated, and then purified by silica gel column chromatography to give the alkynyl-indazole.
  • Note: Alternate catalysts include PdCl2(PPh3)2; Cosolvents include triethylamine and pyrrolidine; When employing bromo-heterocycles, the reaction temperature was increased (80-120° C.).
  • General Procedure C: Multi-component cross-coupling of the alkynyl-indazoles.
  • Figure US20120071535A1-20120322-C00223
  • A mixture of the appropriate alkynyl-indazole (1.0 equiv), aryl-iodide (3.0 equiv), aryl-boronic acid (3.0 equiv), K2CO3 (3.0 equiv), and N,N-dimethylformamide (DMF)/water (2:1, 50 mL/mmol) was degassed with three vacuum/N2 cycles and then heated at 45° C. After 10 min (or when homogenous), a solution of Pd(PhCN)2Cl2 (0.01 equiv) in DMF was added. The reaction was stirred at 45° C. for 4-24 h (until complete by TLC or LCMS), allowed to cool to room temperature, quenched with water, and then extracted with ethyl acetate. The extracts were washed with water, washed with brine, dried, filtered, concentrated, and purified by silica gel chromatography to give the desired tetra-substituted alkene.
  • Note 1: In some instances, all chemicals were simply mixed at room temperature, degassed, and then heated. In other instances, the boronic acid was added last as a DMF/water solution.
  • Note 2: When incomplete conversion of alkynyl-indazole was observed (especially with ortho-substituted aryl-iodides), additional aryl-iodide, aryl-boronic acid, and K2CO3 (1-3 equiv each) were added, and heating was continued for 8-24 h. In some instances, this was repeated multiple times to improve the conversion and yield.
  • General Procedure D: Alternate multi-component cross-coupling of the alkynyl-indazoles.
  • Figure US20120071535A1-20120322-C00224
  • Step 1: Formation of bis(pinacolato)diboryl-alkene
  • Figure US20120071535A1-20120322-C00225
  • A solution of the appropriate alkynyl-indazole (1.0 equiv), bis(pinacolato)diboron (1.01-1.02 equiv), Pt(PPh3)4 (0.0025-0.03 equiv; Note 1), and solvent (2 mL/mmol of dioxane, DME, 2-MeTHF, PhMe, or DMA; Note 2) was degassed with three vacuum/N2 cycles and then heated at 80-120° C. (Note 3) under N2 for 1-8 h (until complete by TLC or LCMS). The reaction was allowed to cool to room temperature and then either 1) taken directly into Step 2; 2) concentrated to give a crude residue [usually a foam]; or 3) concentrated and purified by silica gel chromatography to give the pure bis(pinacolato)diboryl-alkene.
  • Note 1: Most commonly, 0.01 equivalents were utilized. Note 2: Most commonly, 2-MeTHF was utilized. Note 3: Most commonly, reactions were refluxed.
  • Step 2: Cross-coupling of the bis(pinacolato)diboryl-alkene
  • Figure US20120071535A1-20120322-C00226
  • A mixture of bis(pinacolato)diboryl-alkene (1.0 equiv), an appropriate 4-iodoaryl-aldehyde (1.0 equiv), PdCl2(PPh3)2 (0.02-0.1 equiv; Note 1), Cs2CO3 (1.3-3 equiv; Note 2), solvent (4 ml/mmol: dioxane, DME, 2-MeTHF, PhMe, DMA; Note 3 & 4), and water (0-3% v/v; Note 5) was stirred vigorously at 20-40° C. (Note 6) under N2 (Note 7) for 1-24 h (until complete by TLC or LCMS). The reaction was then either 1) taken directly into Step 3; or 2) processed to isolate the 1-aryl-2-(pinacolato)boryl-alkene: [The reaction was diluted with ethyl ether (or ethyl acetate) and washed with water (1-3 times). The aqueous phases were back extracted with ethyl ether (or ethyl acetate). The extracts were combined, dried, filtered, concentrated and then purified by silica gel chromatography].
  • Note 1: Most commonly, 0.1 equivalents were used. Alternate catalysts include PdCl2(dppf). Note 2: Most commonly, 2 or 3 equivalents were used. Water content of the Cs2CO3 affects this reaction, see Note 5. Note 3: Most commonly, 2-MeTHF was used. Note 4: When the bis(pinacolato)diboryl-alkene is brought into this step as a solution from Step 1, solvent (2 mL/mmol) is added to make the final volume of solvent approximately 4 mL/mmol. Note 5: Most commonly, anhydrous Cs2CO3 and anhydrous solvent were used, so 1-2% water (v/v with respect to solvent) was added to the reaction. When the Cs2CO3 and/or solvent were not anhydrous, no water was added. Note 6: Most commonly, reactions were run at room temperature. Note 7: In some instances, this reaction was degassed with three vacuum/N2 cycles.
  • Step 3: Cross-coupling of the 1-aryl-2-(pinacolato)boryl-alkene
  • Figure US20120071535A1-20120322-C00227
  • A mixture of 1-aryl-2-(pinacolato)boryl-alkene (1.0 equiv), an appropriate aryl-halide (1.3-2 equiv; Note 1), PdCl2(PPh3)2 (0.02-0.1 equiv; Note 2), solvent (4 mL/mmol: dioxane, DME, 2-MeTHF, DMSO; Notes 3 & 4), and KOH (3-6M, 5-6 equiv; Note 5) was degassed with three vacuum/N2 cycles and then heated at 80-100° C. (Note 6) under N2 for 1-24 h (until complete by TLC or LCMS). The reaction was allowed to cool to room temperature, diluted with ethyl ether (or ethyl acetate), and washed with water (1-3 times). The aqueous phases were back extracted with ethyl ether (or ethyl acetate). The extracts were combined, dried, filtered, concentrated and then purified by silica gel chromatography to give the desired tetra-substituted alkene.
  • Note 1: Most commonly, 1.5 equivalents of aryl-iodide were used. Note 2: Most commonly, 0.1 equivalents were used. Alternate catalysts include PdCl2(dppf). Note 3: Most commonly, dioxane, DME, or 2-MeTHF was used. Note 4: When the 1-aryl-2-(pinacolato)boryl-alkene is brought into this step directly from Step 2, no additional solvent or PdCl2(PPh3)2 was added. Only the aryl-halide and KOH are added. Note 5: Most commonly, 6 equiv of KOH are used, and the aqueous solution of KOH is 4M or 6M. For compounds with sensitive functionality, K2CO3 (6 equiv, 4M aqueous) is used in place of KOH, and DMSO is used as either the sole solvent or a co-solvent. Note 6: Most commonly, reactions were refluxed.
  • General Procedure E: Olefination of the tetrasubstituted-alkene aryl-aldehydes
  • Figure US20120071535A1-20120322-C00228
  • 1,8-Diazabicyclo[5.4.0]undec-7-ene (DBU, 1.1 equiv) was added dropwise to a mixture of the appropriate aryl-aldehyde (1.0 equiv), triethylphosphonoacetate (1.1-1.3 equiv), lithium chloride (2.0 equiv), and anhydrous acetonitrile (2 mL/mmol) at room temperature. The resulting mixture was stirred for 1-4 h (until complete by TLC or LCMS) and then concentrated. Dichloromethane (or ethyl acetate or ether) was added, and the mixture was washed with water, washed with brine, dried, filtered, concentrated, and purified by silica gel column chromatography to give the desired acrylic ester.
  • Note 1: In some instances, alternate phosphonate reagents were utilized to give the desired acrylic ester. Note 2: Alternate reaction conditions: 1-2 equivalents of phosphonate in THF at −78° C. or 0° C. were treated with n-BuLi or NaH (1-2 equiv). Then aryl-aldehyde (1 equiv) was added, and the reaction was continued at −78° C., 0° C., or room temperature until the reaction was complete by TLC and/or LCMS.
  • General Procedure F: Removal of the tetrahydropyran protecting group from the indazole.
  • Figure US20120071535A1-20120322-C00229
  • A solution of HCl (Note 1) was added to a solution of the THP-protected indazole (1.0 equiv) in ethanol (2-5 mL/mmol; Note 2) at room temperature. The mixture was heated at 70° C. (Note 3) for 2-8 h (until complete by TLC or LCMS), allowed to cool to room temperature, and concentrated to give a crude product that was either carried on directly to the next step or purified by silica gel chromatography.
  • Note 1: Most commonly, 2M HCl in diethyl ether or 1.25M HCl in ethanol were used. Most commonly, the volume of HCl solution used was 10% of the solvent volume. Note 2: Most commonly, the concentration was 5 mL/mmol. In some instances, methanol or isopropanol were used. Note 3: In some instances, the reaction was heated at 80° C. or reflux.
  • General Procedure G: Hydrolysis of the acrylic ester to the acrylic acid.
  • Figure US20120071535A1-20120322-C00230
  • An aqueous solution of LiOH (2-20 equiv; Note 1) was added to a solution of the appropriate ester (1.0 equiv) in ethanol/tetrahydrofuran (1:1, 10 mL/mmol; Note 2) at room temperature, and the mixture was stirred for 4-24 h (until complete by TLC or LCMS). A solution of HCl (1M aqueous) was added until the pH was 3 (Note 3). The mixture was diluted with water and extracted with ethyl acetate (or dichloromethane or ether). The organic layer was washed with water, washed with brine, dried, filtered, concentrated, and purified by silica gel chromatography or preparative-HPLC to give the desired acrylic acid.
  • Note 1: Most commonly, a 2M solution of aqueous LiOH was used, or the LiOH was dissolved in a minimum amount of water. In some instances, NaOH or KOH was used. Note 2: In some instances, a single solvent (ethanol, dioxane, or tetrahydrofuran) was used. Note 3: Alternate work-up procedures have been employed including: i) the use of sat'd NH4Cl in place of aqueous HCl and ii) removal of the organic solvent by rotary evaporation prior to acid quench.
  • Example 28 Preparation of Compound 1: (E)-Ethyl 3-(4-((E)-1-(1H-indazol-5-yl)-2-phenylbut-1-en-1-yl)phenyl)acrylate Step 1: (E)-Ethyl 3-(4-((E)-2-phenyl-1-(1-(tetrahydro-2H-pyran-2-yl)-1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylate
  • Figure US20120071535A1-20120322-C00231
  • A solution of 5-(but-1-yn-1-yl)-1-(tetrahydro-2H-pyran-2-yl)-1H-indazole (2.5 g, 9.83 mmol, Intermediate 3), iodobenzene (6 g, 29.5 mmol), (E)-(4-(3-ethoxy-3-oxoprop-1-en-1-yl)phenyl)boronic acid (6.49 g, 29.5 mmol), K2CO3 (4.08 g, 29.5 mmol), and N,N-dimethylformamide/water (2:1, 492 mL) was degassed with 3 vacuum/N2 cycles and then heated at 45° C. until it was a homogenous solution. A solution of Pd(PhCN)2Cl2 (38 mg, 0.098 mmol) in N,N-dimethylformamide (0.5 mL) was added. The resulting mixture was stirred at 45° C. overnight. Upon completion, the reaction mixture was cooled down to room temperature, quenched with water (500 mL), and extracted with ethyl acetate (3×500 mL). The combined organics were washed with water, washed with brine, dried over sodium sulfate, filtered, and concentrated to give the crude product. This crude material was purified on a silica gel column eluted with 0-50% ethyl acetate in hexanes affording the title compound as off-white foam (3.71 g). LCMS: 423 [(M-THP+H)+H]+.
  • Step 2: (E)-Ethyl 3-(4-((E)-1-(1H-indazol-5-yl)-2-phenylbut-1-en-1-yl)phenyl)acrylate
  • Figure US20120071535A1-20120322-C00232
  • To a solution of (E)-ethyl 3-(4-((E)-2-phenyl-1-(1-(tetrahydro-2H-pyran-2-yl)-1H-indazol-5-yl)but-1-en-1-yl)phenypacrylate (3.5 g, 6.9 mmol) in ethyl alcohol (69 mL), HCl (6 mL, 2M in diethyl ether) was added at room temperature. The resulting mixture was then heated at 70° C. for 2 h. Upon completion, the mixture was cooled down to room temperature and concentrated to give the crude product. This crude material was purified on a silica gel column eluted with 0-100% ethyl acetate in hexanes affording an off-white solid (2.5 g). 1H NMR (300 MHz, DMSO-d6): δ 13.10 (s, 1H), 8.08 (s, 1H), 7.69 (s, 1H), 7.53 (d, 1H), 7.48 (d, 1H), 7.39 (d, 2H), 7.27-7.11 (m, 6H), 6.89 (d, 2H), 6.45 (d, 1H), 4.20 (q, 2H), 2.43 (q, 2H), 1.22 (t, 3H), 0.87 (t, 3H); LCMS: 423 (M+H)+.
  • Example 29 Preparation of Compound 2: (E)-3-(4-((E)-1-(1H-Indazol-5-yl)-2-phenylbut-1-en-1-yl)phenyl)acrylic acid
  • Figure US20120071535A1-20120322-C00233
  • To a solution of (E)-ethyl 3-(4-((E)-1-(1H-indazol-5-yl)-2-phenylbut-1-en-1-yl)phenypacrylate (2.5 g, 5.9 mmol; Compound 1) in THF-EtOH (1:1, 59 mL), an aqueous solution of LiOH (2.8 g, 118 mmol; dissolved in a minimum amount of water) was added at room temperature. The resulting mixture was stirred overnight. The reaction was monitored by LCMS. Upon completion, 1N aqueous HCl was added until pH was 3. Then, the mixture was diluted with water and extracted with ethyl acetate (3×200 mL). The combined organic layers were washed with water, washed with brine, dried over sodium sulfate, filtered, and concentrated to give the crude product. This crude material was purified on a silica gel column eluted with 0-20% methanol in dichloromethane affording the title compound as a pale yellow solid (1.9 g). 1H NMR (300 MHz, DMSO-d6): δ 13.11 (s, 1H), 12.30 (br, 1H), 8.08 (s, 1H), 7.65 (s, 1H), 7.53 (d, 1H), 7.43 (d, 1H), 7.37 (d, 2H), 7.29-7.11 (m, 6H), 6.88 (d, 2H), 6.37 (d, 1H), 2.44 (q, 2H), 0.87 (t, 3H); LCMS: 395 (M+H)+.
  • Compounds 3 to 89 were prepared from alkynyl-indazole intermediates following General Procedures C, F, & G. The alkynyl-indazole intermediates have either i) been described herein or ii) were prepared from known or commercially available halo-indazoles following General Procedures A & B.
  • Compounds 90 to 92 are intermediates from the synthesis of Compounds 3, 12, &13.
  • Example 30 Preparation of Compound 93: (E)-3-(4-((E)-2-(3-Hydroxyphenyl)-1-(1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylic acid
  • Figure US20120071535A1-20120322-C00234
  • A 10-mL recovery flask equipped with a magnetic stir bar, a rubber septum and N2 inlet was charged with (E)-3-(4-((E)-1-(1H-indazol-5-yl)-2-(3-methoxyphenyl)but-1-en-1-yl)phenyl)acrylic acid (30 mg, 0.07 mmol, Compound 5) and DCM (1.4 mL). This solution was cooled down to 0° C. in an ice-bath. Then, BBr3 (88 mg, 0.35 mmol) was added dropwise via a syringe. The reaction mixture was stirred at 0° C. for 1 h. Upon completion, the reaction was quenched with methanol (5 mL) at 0° C. The resulting mixture was concentrated under reduced pressure to give the crude product that was directly purified on a reversed phased C-18 column eluted with 40-100% acetonitrile in water in the presence of 0.1% TFA affording the title compound as an off-white solid (11 mg). 1H NMR (300 MHz, DMSO-d6): δ 13.11 (s, 1H), 12.32 (br, 1H), 9.23 (s, 1H), 8.08 (s, 1H), 7.62 (s, 1H), 7.52 (d, 1H), 7.45-7.35 (m, 3H), 7.12 (d, 1H), 7.00 (t, 1H), 6.90 (d, 2H), 6.59-6.53 (m, 3H), 6.36 (d, 1H), 2.37 (q, 2H), 0.89 (t, 3H); LCMS: 411 (M+H)
  • Example 31 Preparation of Compound 94: (E)-3-(4-((E)-2-(2-Hydroxyphenyl)-1-(1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylic acid
  • Figure US20120071535A1-20120322-C00235
  • A 10-mL recovery flask equipped with a magnetic stir bar, a rubber septum and N2 inlet was charged with (E)-ethyl 3-(4-((E)-1-(1H-indazol-5-yl)-2-(2-methoxyphenyl)but-1-en-1-yl)phenyl)acrylate (145 mg, 0.32 mmol, an intermediate in the synthesis of Compound 12) in DCM (6 mL). This solution was cooled down to −78° C. in an IPA/dry ice-bath. Then, BBr3 (24.1 mg, 0.96 mmol) was added dropwise via a syringe. The reaction mixture was gradually warmed to 0° C. for 1 h. Upon completion, the reaction was quenched with methanol (5 mL) at 0° C. The resulting mixture was concentrated under reduced pressure to give the crude product. Then, this crude product was dissolved in THF-EtOH (1:1, 6 mL), and aqueous LiOH (0.15 g, 6.4 mmol) was added at room temperature. The resulting mixture was stirred at this temperature overnight. Reaction was monitored by LCMS. Upon completion, 1N aqueous HCl was added until pH was 3. Then, the mixture was diluted with water and extracted with ethyl acetate (3×100 mL). The combined organic layers were washed with water (100 mL), washed with brine (50 mL), dried over sodium sulfate, filtered, and concentrated to give the crude product. This crude material was purified on a reversed phase C-18 column eluted with 40-100% acetonitrile in water in the presence of 0.1% TFA affording the title compound. 1H NMR (300 MHz, DMSO-d6): δ 13.07 (s, 1H), 12.34 (br, 1H), 9.33 (br, 1H), 8.08 (d, 1H), 7.65 (s, 1H), 7.53 (d, 1H), 7.40 (d, 1H), 7.32 (d, 2H), 7.15 (dd, 1H), 7.00-6.94 (m, 3H), 6.81-6.76 (m, 2H), 6.57 (dt, 1H), 6.34 (d, 1H), 2.43-2.30 (m, 2H), 0.88 (t, 3H); LCMS: 411 (M+H)+.
  • Compound 95 was prepared following the procedure outlined for Compound 94.
  • Example 32 Preparation of Compound 96: (E)-3-(4-((E)-2-(3-Butoxyphenyl)-1-(1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylic acid Step 1: (E)-Ethyl 3-(4-((E)-2-(3-hydroxyphenyl)-1-(1-(tetrahydro-2H-pyran-2-yl)-1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylate
  • Figure US20120071535A1-20120322-C00236
  • The title compound was prepared from Intermediate 3,3-iodophenol, and (E)-(4-(3-ethoxy-3-oxoprop-1-en-1-yl)phenyl)boronic acid following General Procedure C.
  • Step 2: (E)-Ethyl 3-(4-((E)-2-(3-butoxyphenyl)-1-(1-(tetrahydro-2H-pyran-2-yl)-1H-indazol-5-yl)but-1-en-1-yl)phenypacrylate
  • Figure US20120071535A1-20120322-C00237
  • Potassium carbonate (53 mg, 0.38 mmol) was added to (E)-ethyl 3-(4-((E)-2-(3-hydroxyphenyl)-1-(1-(tetrahydro-2H-pyran-2-yl)-1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylate (101 mg, 0.19 mmol) in CH3CN (1 mL). After stirring for 15 min, iodobutane (24 μL, 0.21 mmol) was added. The reaction was stirred at rt for 15 h. Additional iodobutane (24 μL, 0.21 mmol) was added, and the reaction was stirred at 60° C. for 10 h and then at rt for 48 h. The reaction was diluted with dichloromethane and filtered through celite. The filtrate was concentrated and purified by silica gel chromatography (0-20% EtOAc in hexanes) to give 97 mg of (E)-ethyl 3-(4-((E)-2-(3-butoxyphenyl)-1-(1-(tetrahydro-2H-pyran-2-yl)-1H-indazol-5-yl)but-1-en-1-yl)phenypacrylate as a white foam. 1H NMR (400 MHz, DMSO-d6): δ 8.12 (s, 1H), 7.73 (d, 1H), 7.64 (s, 1H), 7.49 (d, 1H), 7.41 (d, 2H), 7.24 (dd, 1H), 7.12 (t, 1H), 6.91 (d, 2H), 6.76 (d, 1H), 6.67-6.72 (m, 2H), 6.48 (d, 1H), 5.86 (d, 1H), 4.15 (q, 2H), 3.86-3.94 (m, 1H), 3.72-3.80 (m, 3H), 2.38-2.46 (m, 3H), 1.96-2.10 (m, 2H), 1.70-1.82 (m, 1H), 1.52-1.63 (m, 4H), 1.31-1.37 (m, 2H), 1.22 (t, 3H), 0.85-0.92 (m, 6H); LCMS: 495 [(M-THP+H)+ 11]+.
  • Step 3: (E)-3-(4-((E)-2-(3-Butoxyphenyl)-1-(1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylic acid
  • Figure US20120071535A1-20120322-C00238
  • The title compound was prepared from (E)-ethyl 3-(4-((E)-2-(3-butoxyphenyl)-1-(1-(tetrahydro-2H-pyran-2-yl)-1H-indazol-5-yl)but-1-en-1-yl)phenypacrylate following General Procedures F and G. 1H NMR (400 MHz, DMSO-d6): δ 13.11 (bs, 1H), 12.33 (bs, 1H), 8.08 (s, 1H), 7.64 (s, 1H), 7.54 (d, 1H), 7.43 (d, 1H), 7.37 (d, 2H), 7.10-7.17 (m, 2H), 6.91 (d, 2H), 6.75 (d, 1H), 6.66-6.72 (m, 2H), 6.37 (d, 1H), 3.78 (t, 2H), 2.43 (q, 2H), 1.52-1.60 (m, 2H), 1.29-1.38 (m, 2H), 0.85-0.92 (m, 6H); LCMS: 467 (M+H)+.
  • Compound 97 was prepared following the procedure outlined for Compound 96.
  • Example 33 Preparation of Compound 98: (E)-3-(4-((E)-1-(1H-Indazol-5-yl)-2-(2-(methylsulfonyl)phenyl)but-1-en-1-yl)phenyl)acrylic acid Step 1: (E)-Ethyl 3-(4-((E)-2-(2-(methylthio)phenyl)-1-(1-(tetrahydro-2H-pyran-2-yl)-1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylate
  • Figure US20120071535A1-20120322-C00239
  • The title compound was prepared from Intermediate 3,2-iodothioanisole, and (E)-(4-(3-ethoxy-3-oxoprop-1-en-1-yl)phenyl)boronic acid following General Procedure C. 1H NMR (DMSO-d6, 300 MHz): δ 8.14 (s, 1H), 7.75 (d, 1H), 7.72-7.65 (m, 1H), 7.44 (d, 1H), 7.35 (d, 2H), 7.29-7.24 (m, 1H), 7.22-7.11 (m, 3H), 7.09-7.03 (m, 1H), 7.01 (d, 2H), 6.44 (d, 1H), 5.85 (dd, 1H), 4.13 (q, 2H), 3.94-3.83 (m, 1H), 3.80-3.68 (m, 1H), 2.47-2.27 (m, 6H), 2.09-1.93 (m, 2H), 1.83-1.69 (m, 1H), 1.67-1.52 (m, 2H), 1.20 (t, 3H), 0.88 (t, 3H).
  • Step 2: (E)-Ethyl 3-(4-((E)-2-(2-(methylsulfonyl)phenyl)-1-(1-(tetrahydro-2,1-pyran-2-yl)-1H-indazol-5-yl)but-1-en-1-yl)phenypacrylate
  • Figure US20120071535A1-20120322-C00240
  • Potassium peroxymonosulfate (521 mg, 0.85 mmol) was added to a slurry of (E)-ethyl 3-(4-((E)-2-(2-(methylthio)phenyl)-1-(1-(tetrahydro-2H-pyran-2-yl)-1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylate (156 mg, 0.28 mmol) in MeOH:H2O (1:1, 6 mL) at room temperature, and the reaction was stirred overnight. DCM and water were added, and the layers were separated. The aqueous layer was washed with DCM (×2). The organic layers were combined, washed with water, washed with brine, dried over Na2SO4, filtered, and concentrated. The crude material was purified on a silica gel column eluted with 0-50% ethyl acetate in hexane affording the title compound. 1H NMR (DMSO-d6, 300 MHz): δ 8.15 (s, 1H), 7.91 (d, 1H), 7.77-7.71 (m, 2H), 7.49-7.46 (m, 3H), 7.41-7.31 (m, 4H), 7.01 (d, 2H), 6.45 (d, 1H), 5.87 (dd, 1H), 4.12 (q, 2H), 3.92-3.85 (m, 1H), 3.82-3.69 (m, 1H), 2.93 (s, 3H), 2.46-2.27 (m, 2H), 2.09-1.97 (m, 3H), 1.85-1.67 (m, 1H), 1.63-1.51 (m, 2H), 1.18 (t, 3H), 0.83 (t, 3H). LCMS: 501 [(M-THP+H)+H]+.
  • Step 3: (E)-3-(4-((E)-1-(1H-Indazol-5-yl)-2-(2-(methylsulfonyl)phenyl)but-1-en-1-yl)phenyl)acrylic acid
  • Figure US20120071535A1-20120322-C00241
  • The title compound was prepared from (E)-ethyl 3-(4-((E)-2-(2-(methylsulfonyl)phenyl)-1-(1-(tetrahydro-2H-pyran-2-yl)-1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylate following General Procedures F & G. 1H NMR (DMSO-d6, 300 MHz): δ 13.14 (br s, 1H), 12.29 (br s, 1H), 8.11 (d, 1H), 7.92 (dd, 1H), 7.71 (s, 1H), 7.66-7.54 (m, 2H), 7.53-7.44 (m, 2H), 7.42-7.33 (m, 3H), 7.26 (dd, 1H), 7.01 (d, 2H), 6.34 (d, 1H), 2.94 (s, 3H), 2.42-2.30 (m, 2H), 0.83 (t, 3H); LCMS: 473 (M+H)+.
  • Example 34 Preparation of Compound 99: (E)-3-(4-((E)-1-(1H-Indazol-5-yl)-2-phenylbut-1-en-1-yl)phenyl)-2-methylacrylic acid Step 1: (E)-4-(2-Phenyl-1-(1-(tetrahydro-2H-pyran-2-yl)-1H-indazol-5-yl)but-1-en-1-yl)benzaldehyde
  • Figure US20120071535A1-20120322-C00242
  • The title compound was prepared from Intermediate 3, iodobenzene, and (4-formylphenyl)boronic acid following General Procedure C. LCMS: 353 [(M-THP+H)+H]+.
  • Step 2: (E)-Ethyl 2-methyl-3-(4-((E)-2-phenyl-1-(1-(tetrahydro-2H-pyran-2-yl)-1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylate
  • Figure US20120071535A1-20120322-C00243
  • To a suspension of NaH (80 mg, 2 mmol, 60% dispersion in mineral oil) in THF (10 mL) at 0° C., ethyl 2-(diethoxyphosphoryl)propanoate (0.36 g, 1.5 mmol) was added. The reaction was stirred at 0° C. for 1 h, and then a THF solution of (E)-4-(2-phenyl-1-(1-(tetrahydro-2H-pyran-2-yl)-1H-indazol-5-yl)but-1-en-1-yl)benzaldehyde (0.44 g, 1 mmol) was added. The resulting mixture was gradually warmed to room temperature and stirred overnight. The reaction mixture was quenched with saturated ammonium chloride solution and extracted with EtOAc (2×100 mL). Combined organic layers were washed with water, brine, dried over sodium sulfate, filtered and concentrated to give the crude material as pale yellow oil. LCMS: 437 [(M-THP+H)+H]+.
  • Step 3: (E)-3-(4-((E)-1-(1H-Indazol-5-yl)-2-phenylbut-1-en-1-yl)phenyl)-2-methylacrylic acid
  • Figure US20120071535A1-20120322-C00244
  • The title compound was prepared from (E)-ethyl 2-methyl-3-(4-((E)-2-phenyl-1-(1-(tetrahydro-2H-pyran-2-yl)-1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylate following General Procedures F & G. 1H NMR (300 MHz, DMSO-d6): δ 13.11 (s, 1H), 12.55 (br, 1H), 8.08 (d, 1H), 7.65 (s, 1H), 7.53 (d, 1H), 7.42 (d, 1H), 7.21-7.11 (m, 8H), 6.90 (d, 2H), 2.40 (q, 2H), 1.92 (d, 3H), 0.87 (t, 3H); LCMS: 409 (M+H)+.
  • Compounds 100 to 109 were prepared from the appropriate boronic acid or phosphonate following the procedures outlined for Compound 99 or General Procedures C, E, F & G.
  • Example 35 Preparation of Compound 110: (E)-Ethyl 3-(4-((E)-2-(2-chloro-4-fluorophenyl)-1-(1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylate hydrochloride Step 1: (E)-4-(2-(2-Chloro-4-fluorophenyl)-1-(1-(tetrahydro-2H-pyran-2-yl)-1,1-indazol-5-yl)but-1-en-1-yl)benzaldehyde
  • Figure US20120071535A1-20120322-C00245
  • A round-bottom flask equipped with a magnetic stir bar, a reflux condenser, internal thermometer, and a N2 inlet was charged with 5-(but-1-yn-1-yl)-1-(tetrahydro-2H-pyran-2-yl)-1H-indazole (50.0 g, 197 mmol; Intermediate 3), bis(pinacolato)diboron (50.4 g, 199 mmol), and anhydrous 2-methyltetrahydrofuran (393 mL) followed by Pt(PPh3)4 (1.83 g, 1.5 mmol). This mixture was degassed with three vacuum/N2 cycles, heated at 83° C. (internal temperature; oil bath at 95° C.) for 5 h under N2, and then allowed to cool to room temperature. 2-Methyltetrahydrofuran (393 mL), cesium carbonate (128.1 g, 393 mmol), and water (11.8 mL, 1.5% v/v) were added, and the reaction was cooled to 4° C. 4-Iodobenzaldehyde (45.6 g, 197 mmol) and PdCl2(PPh3)2 (6.90 g, 9.8 mmol) were added, and the reaction was degassed with three vacuum/N2 cycles. The mixture was allowed to warm to room temperature and stirred overnight. Aqueous KOH solution (4M, 275 mL, 1100 mmol) and 2-chloro-4-fluoroiodobenzene (70.6 g, 275 mmol) were added. The reaction was degassed with 3 vacuum/N2 cycles, heated at 75° C. (internal temperature; oil bath at 90° C.) for 7 h under N2, and then allowed to cool to room temperature. The layers were separated, and the organic layer was washed with brine (800 mL), dried over sodium sulfate, filtered, and concentrated. The crude product was purified by silica gel chromatography (0-20% ethyl acetate in hexanes) to give the title compound (82.6 g, 7:1 mixture of regioisomers) as a pale yellow foam. Data for major isomer; (E)-4-(2-(2-chloro-4-fluorophenyl)-1-(1-(tetrahydro-2H-pyran-2-yl)-1H-indazol-5-yl)but-1-en-1-yl)benzaldehyde: 1H NMR (300 MHz, DMSO-d6): δ 9.82 (s, 1H), 8.15 (s, 1H), 7.78-7.71 (m, 2H), 7.61 (d, 2H), 7.43-7.27 (m, 3H), 7.15 (m, 3H), 5.86 (dd, 1H), 3.93-3.85 (m, 1H), 3.79-3.68 (m, 1H), 2.44-2.36 (m, 3H), 2.10-1.96 (m, 2H), 1.81-1.67 (m, 1H), 1.63-1.53 (m, 2H), 0.92 (t, 3H); LCMS: 405 [(M-THP+H)+H]+.
  • Step 2: (E)-Ethyl 3-(4-((E)-2-(2-chloro-4-fluorophenyl)-1-(1-(tetrahydro-2H-pyran-2-yl)-1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylate
  • Figure US20120071535A1-20120322-C00246
  • A round-bottom flask equipped with a magnetic stir bar, an addition funnel, and a N2 inlet was charged with (E)-4-(2-(2-chloro-4-fluorophenyl)-1-(1-(tetrahydro-2H-pyran-2-yl)-1H-indazol-5-yl)but-1-en-1-yl)benzaldehyde (82.6 g, 169 mmol), triethylphosphonoacetate (40.6 mL, 203 mmol), lithium chloride (14.5 g, 338 mmol), and anhydrous acetonitrile (338 mL). The reaction was cooled to 0° C. and then degassed with three vacuum/N2 cycles. A solution of DBU (27.8 mL, 186 mmol) in acetonitrile (60 mL) was added dropwise over 35 min, and then the ice water bath was removed. The reaction was stirred at room temperature for 1 h, concentrated, and then partitioned between dichloromethane (250 mL) and H2O (250 mL). The layers were separated, and the organic layer was washed with brine (400 mL), dried over sodium sulfate, filtered, and concentrated. The crude product was passed through a silica gel column (300 g, 20% ethyl acetate in hexanes) and concentrated to give the title compound (89.6 g) as a pale yellow foam. 1H NMR (300 MHz, DMSO-d6): δ 8.14 (s, 1H), 7.75 (d, 1H), 7.50-7.33 (m, 6H), 7.27 (dt, 1H), 7.14 (dt, 1H), 6.95 (d, 2H), 6.48 (d, 1H), 5.86 (dd, 1H), 4.14 (q, 2H), 3.94-3.86 (m, 1H), 3.78-3.70 (m, 1H), 2.45-2.34 (m, 3H), 2.06-1.95 (m, 2H), 1.78-1.67 (m, 1H), 1.62-1.53 (m, 2H), 1.19 (t, 3H), 0.90 (t, 3H); LCMS: 475 [(M-THP+H)+H]+.
  • Step 3: (E)-Ethyl 3-(4-((E)-2-(2-chloro-4-fluorophenyl)-1-(1H-indazol-5-yl)but-1-en-1-yl)phenypacrylate hydrochloride
  • Figure US20120071535A1-20120322-C00247
  • A round-bottom flask equipped with a magnetic stir bar was charged with (E)-ethyl 3-(4-((E)-2-(2-chloro-4-fluorophenyl)-1-(1-(tetrahydro-2H-pyran-2-yl)-1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylate (255.9 g, 457.8 mmol) and a solution of HCl (732 mL, 1.25 M in ethyl alcohol). The reaction was heated at 80° C. for 2.5 h, allowed to cool to room temperature, and then concentrated to an orange gel. tert-Butyl methyl ether (2.3 L) was added. After stirring for 5 min, solids began to precipitate. The mixture was stirred at room temperature for 2 h and then filtered. The solids were washed with MTBE (700 mL) and dried to give the title compound (193 g) as an off-white solid. 1H NMR (DMSO-d6): δ 8.11 (s, 1H), 7.69 (s, 1H), 7.57-7.50 (m, 2H), 7.45-7.33 (m, 4H), 7.21-7.10 (m, 2H), 6.96 (d, 2H), 6.48 (d, 1H), 4.14 (q, 2H), 2.38 (q, 2H), 1.19 (t, 3H), 0.90 (t, 3H); LCMS: 475 (M+H)+.
  • Example 36 Preparation of Compound III: (E)-3-(4-0E)-2-(2-Chloro-4-fluorophenyl)-1-(1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylic acid
  • Figure US20120071535A1-20120322-C00248
  • A round-bottom flask equipped with a magnetic stir bar was charged with (E)-ethyl 3-(4-((E)-2-(2-chloro-4-fluorophenyl)-1-(1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylate hydrochloride (198.5 g, 388 mmol; Compound 110) and ethyl alcohol (517 mL). A solution of LiOH (27.9 g, 1164 mmol) in water (388 mL) was added, and the mixture was stirred at room temperature overnight. The ethyl alcohol was removed by rotary evaporation, and the remaining solution was cooled to 0° C. and acidified with 2M aqueous HCl to pH 3. Dichloromethane (500 mL) was added, the mixture was stirred, and then the layers were separated. The organic layer was washed with water, washed with brine, dried over sodium sulfate, filtered, and concentrated. The crude product was passed through a silica column (800 g, 5% MeOH in DCM) and concentrated. The product was then dissolved in DCM (400 mL), and acetonitrile (500 mL) was added. Approximately 200 mL of DCM was removed by rotary evaporation (solids began to precipitate). Acetonitrile (550 mL) was added followed by water (25 mL). The mixture was stirred at room temperature for 2 h. The solvent was decanted, and then acetonitrile:DCM (10:1; 550 mL) was added. The mixture stirred at room temperature for 1.5 h, the solvent was again decanted, and then acetonitrile:DCM (10:1; 550 mL) was added. The mixture was again stirred at room temperature for 1.5 h and then filtered. The solids were resuspended in acetonitrile:DCM (10:1; 550 mL), stirred at room temperature for 1.5 h, filtered, and washed to give the title compound (123.9 g) as an off-white powder. 1H NMR (DMSO-d6): δ 13.12 (s, 1H), 12.34 (br, 1H), 8.11 (d, 1H), 7.69 (s, 1H), 7.56 (d, 1H), 7.44-7.33 (m, 5H), 7.21-7.10 (m, 2H), 6.96 (d, 2H), 6.38 (d, 1H), 2.34 (q, 2H), 0.90 (t, 3H); LCMS: 447 (M+H)+.
  • Example 37 Preparation of Compound 112: (E)-Ethyl 3-(4-((E)-2-(2,4-dichlorophenyl)-1-(1H-indazol-5-yl)but-1-en-1-yl)phenypacrylate Step 1: (E)-4-(2-(2,4-Dichlorophenyl)-1-(1-(tetrahydro-2H-pyran-2-yl)-1H-indazol-5-yl)but-1-en-1-yl)benzaldehyde
  • Figure US20120071535A1-20120322-C00249
  • A round-bottom flask equipped with a magnetic stir bar, a reflux condenser, and a N2 inlet was charged with 5-(but-1-yn-1-yl)-1-(tetrahydro-2H-pyran-2-yl)-1H-indazole (20.0 g, 78.6 mmol; Intermediate 3), bis(pinacolato)diboron (20.17 g, 79.4 mmol), tetrakis(triphenylphosphine)platinum (0) (0.98 g, 0.8 mmol), and anhydrous 1,4-dioxane (160 mL). This mixture was degassed with three vacuum/N2 cycles and refluxed for 4 h. The solution was then allowed to cool to room temperature, and 4-iodobenzaldehyde (18.25 g, 78.6 mmol), trans-dichloro(triphenylphosphine)palladium (II) (5.52 g, 7.9 mmol), cesium carbonate (51.24 g, 157.3 mmol), and 1,4-dioxane (160 mL) were added. This mixture was degassed with three vacuum/N2 cycles, and then water (4.7 mL) was added. This mixture was stirred at room temperature for 6 h. 2,4-Dichloroiodobenzene (12.8 mL, 94.4 mmol) and 6M aqueous KOH (62.9 mL) were added, and the mixture was degassed with three vacuum/N2 cycles and refluxed for 4 h. Upon completion, the reaction mixture was filtered through a Celite/silica pad and washed with EtOAc. The filtrate was washed with water (600 mL), washed with brine (300 mL), dried over sodium sulfate, filtered, and concentrated. The crude product was purified by silica gel chromatography (0-20% ethyl acetate in hexanes) to give the title compound (27.2 g, 7:1 mixture of regioisomers) as a yellow foam. Data for major regioisomer: 1H NMR (400 MHz, DMSO-d6): δ 9.83 (s, 1H), 8.16 (s, 1H), 7.77 (d, 1H), 7.73 (s, 1H), 7.65 (d, 2H), 7.53 (d, 1H), 7.41-7.36 (m, 2H), 7.31-7.28 (m, 1H), 7.17 (d, 2H), 5.86 (dd, 1H), 3.92-3.86 (m, 1H), 3.78-3.71 (m, 1H), 2.47-2.38 (m, 3H), 2.10-1.96 (m, 2H), 1.81-1.71 (m, 1H), 1.64-1.58 (m, 2H), 0.94 (t, 3H); LCMS: 421 [(M-THP+H)+H]+.
  • Step 2: (E)-Ethyl 3-(4-((E)-2-(2,4-dichlorophenyl)-1-(1-(tetrahydro-2H-pyran-2-yl)-1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylate
  • Figure US20120071535A1-20120322-C00250
  • A round-bottom flask equipped with a magnetic stir bar, a rubber septum, and a N2 inlet was charged with (E)-4-(2-(2,4-dichlorophenyl)-1-(1-(tetrahydro-2H-pyran-2-yl)-1H-indazol-5-yl)but-1-en-1-yl)benzaldehyde (26.7 g, 52.8 mmol), triethylphosphonoacetate (12.7 mL, 63.4 mmol), lithium chloride (4.53 g, 105.7 mmol), and anhydrous acetonitrile (106 mL). A solution of DBU (8.7 mL, 58.1 mmol) in ACN (27 mL) was slowly added dropwise via addition funnel. The resulting mixture was stirred at room temperature for 4 h. Upon completion, the reaction was concentrated and redissolved in DCM. This solution was washed with, water (300 mL), washed with brine (250 mL), dried over sodium sulfate, filtered and concentrated. The crude product was purified by silica gel chromatography (0-20% ethyl acetate in hexanes) to give the title compound (29.0 g) as a pale yellow foam. 1H NMR (400 MHz, DMSO-d6): δ 8.14 (s, 1H), 7.75 (d, 1H), 7.72 (s, 1H), 7.54 (d, 1H), 7.48 (d, 1H), 7.43 (d, 2H), 7.37-7.35 (m, 2H), 7.29-7.26 (m, 1H), 6.97 (d, 2H), 6.48 (d, 1H), 5.86 (dd, 1H), 4.14 (q, 2H), 3.91-3.86 (m, 1H), 3.77-3.71 (m, 1H), 2.48-2.35 (m, 3H), 2.06-1.96 (m, 2H), 1.78-1.71 (m, 1H), 1.62-1.55 (m, 2H), 1.22 (t, 3H), 0.90 (t, 3H); LCMS: 491 [(M-THP+H)+H]+.
  • Step 3: (E)-Ethyl 3-(4-((E)-2-(2,4-dichlorophenyl)-1-(1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylate
  • Figure US20120071535A1-20120322-C00251
  • A solution of HCl (5.0 mL, 2.0 M in diethyl ether) was added to a solution of (E)-ethyl 3-(4-((E)-2-(2,4-dichlorophenyl)-1-(1-(tetrahydro-2H-pyran-2-yl)-1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylate (3.0 g, 5.2 mmol) in ethyl alcohol (25 mL) at room temperature. The resulting mixture was heated at 70° C. for 2 h. Upon completion, the mixture was cooled down to room temperature and concentrated to give a pale yellow solid. This crude material was dissolved in DCM and washed with water (50 mL), washed with brine (50 mL), dried over sodium sulfate, filtered, and concentrated. The crude product was purified by silica gel chromatography (0-100% ethyl acetate in hexanes) to give the title compound (2.37 g) as a pale yellow solid. 1H NMR (400 MHz, DMSO-d6): δ 13.17 (s, 1H), 8.11 (s, 1H), 7.69 (s, 1H), 7.58-7.52 (m, 2H), 7.48 (d, 1H), 7.43 (d, 2H), 7.36-7.32 (m, 2H), 7.19 (dd, 1H), 6.97 (d, 2H), 6.49 (d, 1H), 4.15 (q, 2H), 2.39 (q, 2H), 1.22 (t, 3H), 0.90 (t, 3H); LCMS: 491 (M+H)+.
  • Example 38 Preparation of Compound 113: (E)-3-(4-0E)-2-(2,4-Dichlorophenyl)-1-(1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylic acid
  • Figure US20120071535A1-20120322-C00252
  • A solution of LiOH (0.23 g, 9.6 mmol) in water (3.2 mL) was added to a solution of (E)-ethyl 3-(4-((E)-2-(2,4-dichlorophenyl)-1-(1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylate (2.37 g, 4.8 mmol; Compound 112) in EtOH (20 mL) at room temperature. The resulting mixture was stirred overnight. Upon completion, 1N aqueous HCl was added until the pH was 3. The mixture was diluted with water and extracted with ethyl acetate (2×25 mL). The combined organic layers were washed with water (50 mL), washed with brine (50 mL), dried over sodium sulfate, filtered, and concentrated. The crude product was purified on a preparative reversed-phased HPLC column using 80-95% acetonitrile in water in the presence of 0.1% TFA to give the title compound (1.3 g) as an off-white solid. 1H NMR (400 MHz, DMSO-d6): δ 13.11 (s, 1H), 12.36 (br, 1H), 8.11 (d, 1H), 7.69 (s, 1H), 7.57-7.53 (m, 2H), 7.44-7.35 (m, 5H), 7.19 (dd, 1H), 6.97 (d, 2H), 6.39 (d, 1H), 2.39 (q, 2H), 0.90 (t, 3H); LCMS: 463 (M+H)+.
  • Compounds 114 to 139 were prepared from alkynyl-indazole intermediates following General Procedures D, E, F, & G. The alkynyl-indazole intermediates have either i) been described herein or ii) were prepared from known or commercially available halo-indazoles following General Procedures A & B.
  • Example 39 Preparation of Intermediate 45: (E)-Ethyl 3-(44(Z)-1-(1-(tetrahydro-2H-pyran-2-yl)-1H-indazol-5-yl)-2-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)but-1-en-1-yl)phenypacrylate Step 1: (E)-Ethyl 3-(4-iodophenyl)acrylate
  • Figure US20120071535A1-20120322-C00253
  • N-Iodosuccinimide (2.5 g, 11 mmol) was added to a suspension of 4-(E-3-ethoxy-3-oxo-1-propen-1-yl)phenylboronic acid (2.2 g, 10 mmol) in CH3CN (50 mL) at room temperature. The reaction was covered with foil, stirred for ˜26 hours, and then diluted with EtOAc. The resulting mixture was washed with water (2×100 mL), washed with sodium thiosulfate (100 mL), dried (MgSO4), filtered, and concentrated. The crude material was purified by silica gel chromatography (0-50% EtOAc in hexanes) to give 2.6 g of (E)-ethyl 3-(4-iodophenyl)acrylate as a yellow oil. 1H NMR (400 MHz, DMSO-d6): δ 7.79 (d, 2H), 7.60 (d, 1H), 7.53 (d, 2H), 6.68 (d, 1H), 4.19 (q, 2H), 1.26 (t, 3H); LCMS: 303 (M+H)+.
  • Step 2: (E)-Ethyl 3-(44(Z)-1-(1-(tetrahydro-2H-pyran-2-yl)-1H-indazol-5-yl)-2-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)but-1-en-1-yl)phenyl)acrylate
  • Figure US20120071535A1-20120322-C00254
  • The title compound was be prepared from Intermediate 3 and (E)-ethyl 3-(4-iodophenyl)acrylate following General Procedure D, Steps 1-2. 1H NMR (400 MHz, DMSO-d6): δ 8.09 (s, 1H), 7.70 (d, 1H), 7.58-7.63 (m, 3H), 7.49 (s, 1H), 7.11 (d, 3H), 6.58 (d, 1H), 5.84 (dd, 1H), 4.18 (q, 2H), 3.86-3.91 (m, 1H), 3.70-3.77 (m, 1H), 2.36-2.48 (m, 1H), 2.08-2.15 (m, 2H), 1.95-2.08 (m, 2H), 1.70-1.81 (m, 1H), 1.56-1.62 (m, 2H), 1.25 (t, 3H), 1.12 (s, 12H), 1.01 (t, 3H); LCMS: 473 [(M-THP+H)+H]+.
  • Compounds 140 to 144 were prepared from Intermediate 45 following General Procedures D (Step 3; K2CO3 modification), F, & G.
  • Example 40 Preparation of Compound 145: (E)-3-(4-((E)-2-(2-Chlorophenyl)-1-(1-methyl-1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylic acid
  • Figure US20120071535A1-20120322-C00255
  • To a mixture of (E)-3-(4-((E)-2-(2-chlorophenyl)-1-(1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylic acid (80 mg, 0.19 mmol; Compound 14) and Cs2CO3 (0.15 g, 0.46 mmol) in DMF (3.8 mL) at room temperature, iodomethane (65 mg, 0.46 mmol) was added. The mixture was stirred at room temperature overnight, diluted with water, extracted with EtOAc, and concentrated to give the (E)-methyl 3-(4-((E)-2-(2-chlorophenyl)-1-(1-methyl-1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylate. This residue was redissolved in THF-MeOH (3.8 mL) and an aqueous solution of LiOH (89 mg, 3.7 mmol; dissolved in a minimum amount of water) was added at room temperature. The reaction mixture was stirred overnight, quenched with 1N HCl, extracted with EtOAc, dried over sodium sulfate, filtered and concentrated to give the crude material. This crude product was purified on a RP-C18 column using 50-100% acetonitrile in water in the presence of 0.1% TFA to afford the title compound. 1H NMR (300 MHz, DMSO-d6): δ 12.28 (s, 1H), 8.07 (s, 1H), 7.67-7.64 (m, 2H), 7.48-7.11 (m, 8H), 6.95 (d, 2H), 6.35 (d, 1H), 4.05 (s, 3H), 2.36 (q, 2H), 0.90 (t, 3H). LCMS: 443 (M+H)+.
  • Example 41 Preparation of Compound 146: (E)-3-(4-0E)-2-Cyclobutyl-1-(1-methyl-1H-indazol-5-yl)-2-phenylvinyl)phenyl)acrylic acid Step 1: (E)-Ethyl 3-(4-((E)-2-cyclobutyl-1-(1-methyl-1H-indazol-5-yl)-2-phenylvinyl)phenyl)acrylate
  • Figure US20120071535A1-20120322-C00256
  • Iodomethane (80 mg, 0.84 mmol) was added to a mixture of (E)-ethyl 3-(4-((E)-2-cyclobutyl-1-(1H-indazol-5-yl)-2-phenylvinyl)phenypacrylate (0.25 g, 0.56 mmol; intermediate in the preparation of Compound 84), K2CO3 (0.12 g, 0.84 mmol), and DMF (5.6 mL) at room temperature. The resulting mixture was stirred overnight, diluted with water, and extracted with EtOAc. The extract was washed with water, washed with brine, dried over sodium sulfate, filtered, concentrated, and then purified on a silica gel column using 0-50% EtOAc in hexanes to afford the title compound. LCMS: 463 (M+H)+.
  • Step 2: (E)-3-(4-0E)-2-Cyclobutyl-1-(1-methyl-1H-indazol-5-yl)-2-phenylvinyl)phenyl)acrylic acid
  • Figure US20120071535A1-20120322-C00257
  • The title compound was prepared from (E)-ethyl 3-(4-((E)-2-cyclobutyl-1-(1-methyl-1H-indazol-5-yl)-2-phenylvinyl)phenyl)acrylate as described in General Procedure G. 1H NMR (400 MHz, DMSO-d6): δ 12.26 (s, 1H), 8.06 (s, 1H), 7.68-7.61 (m, 2H), 7.37 (d, 1H), 7.31-7.12 (m, 8H), 6.92 (d, 2H), 6.36 (d, 1H), 4.09 (s, 3H), 3.46-3.39 (m, 1H), 1.84-1.76 (m, 4H), 1.63-1.52 (m, 1H), 1.37-1.32 (m, 1H); LCMS: 435 (M+H)+.
  • Compound 147 was prepared following the procedure outlined for Compound 146.
  • Example 42 Preparation of Compound 148: (E)-3-(4-((E)-2-(2-Chloro-4-fluorophenyl)-1-(1-(difluoromethyl)-1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylic acid
  • Figure US20120071535A1-20120322-C00258
  • A solution of (E)-ethyl 3-(4-((E)-2-(2-chloro-4-fluorophenyl)-1-(1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylate (105, mg, 0.22 mmol; freebase of Compound 110) in DMF (1 mL) was added to a suspension of sodium hydride (11 mg, 0.27 mmol) in DMF (1 mL). The mixture was stirred at room temperature for 1 h, and then difluoroiodomethane was bubbled in for 10 mM The reaction mixture was heated at 80° C. for 3 h and cooled to room temperature. Difluoroiodomethane was bubbled in for additional 10 mM, and the mixture was heated for an additional 1.5 h. The reaction mixture was cooled to room temperature, diluted with ethyl acetate (50 mL), washed (2×25 mL H2O), dried (Na2SO4), and concentrated under reduced pressure. The crude material was purified on a silica gel column to yield a mixture containing the desired intermediate. LCMS: 525(M+H)+. Following General Procedure G, this intermediate gave the title compound. 1H NMR (300 MHz, DMSO-d6): δ 8.91 (s, 1H), 8.17 (t, 1H), 7.77 (s, 1H), 7.74 (d, 1H), 7.45-7.35 (m, 5H), 7.20 (dd, 1H), 7.15 (dt, 1H), 7.00 (d, 2H), 6.38 (d, 2H), 2.41 (q, 2H), 0.92 (t, 3H); LCMS: 497 (M+H)+.
  • Example 43 3×ERE MCF-7 Reporter Assay
  • MCF7 cells were maintained in RPMI 1640 supplemented with 10% FCS. Transcriptional assays were performed by seeding 100 μL of cells at a density of 250,000 cells/mL into 96-well cell culture plates in RPMI 1640 supplemented with 10% charcoal stripped serum and allowed to attach overnight. Cells were transiently transfected using Lipofectin (Life Technologies) according to the manufacturer's protocol. Triplicate transfections were performed using 300 ng 3×ERE-TK-Luc (reporter vector), 50 ng CMVpRL (normalization vector), and 130 ng pCMX (filler DNA). Transfected cells were incubated overnight then treated with ligand. For ER agonist assays, the compounds were serially diluted and 50 μL of compound plus RPMI 1640 supplemented with charcoal stripped serum was added to the cells. For ER antagonist assays, the compounds were serially diluted and 50 μL of compound with RPMI plus 17β-estradiol supplemented with charcoal stripped serum were added to the cells. The final 17β-estradiol concentration used in the antagonist assays was 0.1 nM. Following 24 hour incubation the medium was removed and the cells were lysed in 40 μL of lysis buffer (25 mM Tris Phosphate, 2 mM CDTA, 10% Glycerol, 0.5% Triton X-100, 2 mM DTT). Firefly luciferase activity was measured immediately following the addition of 40 μL luciferase buffer (20 mM tricine, 0.1 mM EDTA, 1.07 mM (MgCO3)4 Mg(OH)2.5H2O, 2.67 mM MgSO4, 33.3 mM DTT, 270 μM Coenzyme A, 470 μM luciferin, 530 μM ATP). Renilla luciferase was measured following the addition of 40 μL colelenterazine buffer (1.1 M NaCl, 2.2 mM Na2EDTA, 0.22 M KxPO4 (pH 5.1), 0.44 mg/mL BSA, 1.3 mM NaN3, 1.43 μM coelenterazine, final pH adjusted to 5.0).
  • Example 44 Breast Cancer Cell Viability Assays
  • MCF-7 cells were adjusted to a concentration of 20,000 cells per mL in RPMI containing 10% FBS and 20 mM HEPES. 16 microliters of the cell suspension (320 cells) was added to each well of a 384 well plate, and the cells were incubated overnight to allow the cells to adhere. The following day an eleven point, serial semilog dilution of each compound was added to the cells in 16 μL at a final concentration ranging from 0.3-0.000003 μM. After 5 days' compound exposure, 16 nt of CellTiter-GLo (Promega, Madison Wis.) was added to the cells the relative luminescence units (RLUs) of each well was determined CellTiter-Glo added to 32 μL of medium without cells was used to obtain a background value. The Percent viability of each sample was determined as follows: (RLU sample-RLU background/RLU untreated cells-RLU background)×100=% viability.
  • Viability effects in additional ER+ breast cancer cell lines, including BT474, CAMA1, MDA-MB-361, ZR-75-1, T47D, can be profiled in assays similar to Example 44.
  • Example 45 Breast Cancer Cell ER-α in Cell Western Assay (SP1)
  • MCF-7 cells were adjusted to a concentration of 200,000 cells per mL in RPMI containing 10% charcoal-stripped FBS and 20 mM HEPES. 16 microliters of the cell suspension (3200 cells) was added to each well of a poly-D-lysine 384 well plate, and the cells were incubated overnight to allow the cells to adhere. The following day an eleven point, serial semilog dilution of each compound was added to the cells in 16 μL at a final concentration ranging from 0.3-0.000003 μM. At 4 or 24 hr post compound addition, the cells were fixed (10% formalin in PBS) for 20 minutes. Cells were permeablized in PBS 0.1% Triton and blocked with LICOR blocking buffer (50 μl/well, 90′). The wells were then incubated overnight at 4° C. with SP1 rabbit monoclonal Ab (Thermo Scientific) diluted 1:1000 in LICOR blocking buffer/0.1% Tween-20. Wells which were treated with blocking buffer with Tween but no antibody were used as a background control. Wells were washed with 0.1% Tween-20/PBS and then incubated in goat anti-rabbit IRDye™ 800CW (LICOR Inc.; 1:1000) and DRAQ5 DNA dye (1:2000 for 2 mM stock) diluted in LICOR blocking buffer containing 0.1% Tween-20 and 0.01% SDS for 60 minutes. Cells were washed (50 μl/well, 5′ each) in 0.1% Tween-20/PBS. Plates were scanned on a LICOR Odyssey infrared imaging system. Integrated intensities in the 800 nm channel and 700 nm channel were measured to determine levels of ER and DNA respectively. Percent ER levels were determined as follows: (Integrated intensity 800 nm sample/integrated intensity 700 nm sample)/(Integrated intensity 800 nm untreated cells/integrated intensity 700 nm untreated cells)×100=% ER levels.
  • Effects on steady state levels of ER-α in additional ER+ breast cancer cell lines, including BT474, CAMA1, MDA-MB-361, ZR-75-1, T47D, can be profiled in assays similar to Example 45.
  • Illustrative biological data for representative compounds disclosed herein is presented in the following table:
  • TABLE 7
    MCF7 ER-α In ER-α In
    MCF7 Viability Cell Western Cell Western
    Viability Assay Max Assay (SP1) Assay (SP1)
    Compound Assay IC50 Response IC50 Max Response
    1 A + A ++
    2 A ++ A ++
    3 A ++ A ++
    4 A ++ A ++
    5 A ++ A ++
    6 B ++ A ++
    7 A ++ A ++
    8 B ++ A ++
    9 A ++ A ++
    10 A ++ A ++
    11 A ++ A ++
    12 A ++ A ++
    13 A ++ A ++
    14 A ++ A ++
    15 A ++ A ++
    16 A ++ A ++
    17 A ++ A ++
    18 A ++ A ++
    19 A ++ A ++
    20 A ++ A ++
    21 A ++ A ++
    22 A ++ A ++
    23 A ++ A ++
    24 A ++ A ++
    25 A ++ A ++
    26 A ++ A ++
    27 A ++ A ++
    28 A ++ A ++
    29 A ++ A ++
    30 A ++ A ++
    31 A ++ A ++
    32 A ++ A ++
    33 A ++ A ++
    34 A ++ A ++
    35 A ++ A ++
    36 A ++ A ++
    37 A ++ A ++
    38 B ++ B ++
    39 B ++ B ++
    40 B ++ B ++
    41 A ++ A ++
    42 A ++ A ++
    43 A ++ A ++
    44 A ++ A ++
    45 A ++ A ++
    46 B + B ++
    47 A ++ A ++
    48 A ++ A ++
    49 A ++ A ++
    50 A ++ A ++
    51 A ++ A ++
    52 A ++ A ++
    53 A ++ A ++
    54 A ++ A ++
    55 A ++ A ++
    56 A ++ A ++
    57 A ++ A ++
    58 A ++ A ++
    59 A ++ A ++
    60 A ++ A ++
    61 A ++ A ++
    62 A ++ A ++
    63 A ++ A ++
    64 A ++ A ++
    65 A ++ A ++
    66 A ++ A ++
    67 A ++ A ++
    68 A ++ A ++
    69 A ++ A ++
    70 A ++ A ++
    71 A ++ A ++
    72 A ++ A ++
    73 A ++ A ++
    74 A ++ A ++
    75 A ++ A ++
    76 A ++ A ++
    77 A ++ A ++
    78 A ++ A ++
    79 A ++ A ++
    80 B ++ A ++
    81 B ++ A ++
    82 A ++ A ++
    83 A ++ A ++
    84 A ++ A ++
    85 A ++ A ++
    86 A ++ A ++
    87 A ++ A ++
    88 A ++ A ++
    89 A ++ A ++
    90 A + A ++
    91 A +
    92 A +
    93 A ++ A ++
    94 B ++ A ++
    95 A ++ A ++
    96 A ++ A ++
    97 A ++ A ++
    98 A ++ A ++
    99 A ++ A ++
    100 A ++ A ++
    101 A ++ A ++
    102 A ++ A ++
    103 B ++ A ++
    104 B ++ A ++
    105 A ++ A ++
    106 A ++ A ++
    107 B ++ A ++
    108 A ++ A ++
    109 A ++ A ++
    110 A + A ++
    111 A ++ A ++
    112 A + A ++
    113 A ++ A ++
    114 A ++ A ++
    115 A ++ A ++
    116 A ++ A ++
    117 A ++ A ++
    118 A ++ A ++
    119 A ++ A ++
    120 A ++ A ++
    121 A ++ A ++
    122 A ++ A ++
    123 A ++ A ++
    124 A ++ A ++
    125 A ++ A ++
    126 A ++ A ++
    127 B + B +
    128 A ++ A ++
    129 A ++ A ++
    130 A ++ A ++
    131 B ++ B ++
    132 A ++ A ++
    133 B + B ++
    134 A ++ A ++
    135 A ++ A ++
    136 A ++ A ++
    137 A ++ A ++
    138 A ++ A ++
    139 A ++ A ++
    140 A ++ A ++
    141 B ++ A ++
    142 A ++ A ++
    143 B ++ A ++
    144 A ++ A ++
    145 A ++ A ++
    146 A ++ A ++
    147 A ++ A ++
    148 A ++ A ++
    A = single IC50 ≦ 100 nM;
    B = single IC50 > 100 nM;
    + = a single % value < 40%;
    ++ = a single % value ≧ 40%
  • Example 46 Ishikawa Uterine Cell Alkaline Phosphatase Assay
  • Subconfuent Ishikawa cells in a T225 are incubated 24 hours in an estrogen free basal medium (EFBM) consisting of DMEM:Ham's F-12 50:50 phenol red free basal medium containing 5% Charcoal Dextran treated FBS and 20 mM HEPES. Cells are plated the following day in EFBM in clear 384 well plates at a concentration of 2.5×105 cells per mL, 16 μl per well (4000 cells per well). A 12 point semilog dilution of each compound is carried out in DMSO and subsequently diluted in EFBM. An equal volume of compound in EFBM is added immediately after plating cells, and the cells are incubated for 3 days. The cells are fixed with 5% formalin, and rinsed with PBS. Alkaline Phosphatase substrate 4-Nitrophenyl phosphate disodium salt hexahydrate is added to a solution containing 2 mM MgCl2, 1 M diethanolamine, and adjusted to pH 9.0. The substrate solution is added to the cell cultures (16 μL per well), and OD405 is measured in a multiwall plate spectrophotometer when the optical density at 405 nm wavelength of cells treated with 17β-estradiol in the concentration range of 1-30 nM reaches 1.0-1.2 absorbance units. Cells treated with DMSO alone serve as a background control. Percent activity in background subtracted samples is measured as follows: % activity=OD405 sample/OD405 max of 17/3-estradiol treated cells×100.
  • Example 47 Ovarian Cancer Cell Viability Assays
  • BG-1, cells were adjusted to a concentration of 20,000 cells per mL in RPMI containing 10% FBS and 20 mM HEPES. 16 microliters of the cell suspension (320 cells) was added to each well of a 384 well plate, and the cells were incubated overnight to allow the cells to adhere. The following day an eleven point, serial semilog dilution of each compound was added to the cells in 16 μL at a final concentration ranging from 0.3-0.000003 μM. After 5 days' compound exposure, 16 μL of CellTiter-GLo (Promega, Madison Wis.) was added to the cells the relative luminescence units (RLUs) of each well was determined CellTiter-Glo added to 32 μL of medium without cells was used to obtain a background value. The Percent viability of each sample was determined as follows: (RLU sample-RLU background/RLU untreated cells-RLU background)×100=% viability.
  • Viability effects in additional ER+ ovarian cancer cell lines, including A1847, SKOV3, SW626, A2780, can be profiled in assays similar to Example 47.
  • Example 48 Ovarian Cancer Cell ER-α in Cell Western Assay
  • BG-1 cells were adjusted to a concentration of 200,000 cells per mL in RPMI containing 10% charcoal-stripped FBS and 20 mM HEPES. 16 microliters of the cell suspension (3200 cells) was added to each well of a poly-D-lysine 384 well plate, and the cells were incubated overnight to allow the cells to adhere. The following day an eleven point, serial semilog dilution of each compound was added to the cells in 16 μL at a final concentration ranging from 0.3-0.000003 μM. At 4 or 24 hr post compound addition, the cells were fixed (10% formalin in PBS) for 20 minutes. Cells were permeablized in PBS 0.1% Triton and blocked with LICOR blocking buffer (50 μl/well, 90′). The wells were then incubated overnight at 4° C. with ER1D5 (Santa Cruz Biotechnology) diluted 1:100 in LICOR blocking buffer/0.1% Tween-20. Wells which were treated with blocking buffer with Tween but no antibody were used as a background control. Wells were washed with 0.1% Tween-20/PBS and then incubated in goat anti-mouse IRDye™ 800CW (LICOR Inc.; 1:1000) and DRAQ5 DNA dye (1:2000 for 2 mM stock) diluted in LICOR blocking buffer containing 0.1% Tween-20 and 0.01% SDS for 60 minutes. Cells were washed (50 μl/well, 5′ each) in 0.1% Tween-20/PBS. Plates were scanned on a LICOR Odyssey infrared imaging system. Integrated intensities in the 800 nm channel and 700 nm channel were measured to determine levels of ER and DNA respectively. Percent ER levels were determined as follows: (Integrated intensity 800 nm sample/integrated intensity 700 nm sample)/(Integrated intensity 800 nm untreated cells/integrated intensity 700 nm untreated cells)×100=% ER levels.
  • Effects on steady state levels of ER-α in additional ER+ ovarian cancer cell lines, including A1847, SKOV3, SW626, A2780, can be profiled in assays similar to Example 48.
  • Other cancer cell lines contemplated for testing compounds described herein include: ER-positive endometrial cell lines (Ishikawa, ECC1, HEC-1, EnCa-101) and ER-positive cervical cell lines (Caski, HeLa, SiHa).
  • Example 49 Breast Cancer Model; Xenograft Assay (MCF-7)
  • Time release pellets containing 0.72 mg 17-β Estradiol were subcutaneously implanted into nu/nu mice. MCF-7 cells were grown in RPMI containing 10% FBS at 5% CO2, 37° C. Cells were spun down and re-suspended in 50% RPMI (serum free) and 50% Matrigel at 1×107 cells/mL. MCF-7 cells were subcutaneously injected (100 μL/animal) on the right flank 2-3 days post pellet implantation. Tumor volume (length×width2/2) was monitored bi-weekly. When tumors reached an average volume of ˜200 mm3 animals were randomized and treatment was started. Animals were treated with Vehicle or Compound daily for 4 weeks. Tumor volume and body weight were monitored bi-weekly throughout the study. At the conclusion of the treatment period, plasma and tumor samples were taken for pharmacokinetic and pharmacodynamic analyses, respectively.
  • Example 50 Tamoxifen-resistant Breast Cancer Model; Xenograft Assay (MCF-7 derivative)
  • Female nu/nu mice (with supplemental 17-(Estradiol pellets; 0.72 mg; 60 day slow release) bearing MCF-7 tumors (mean tumor volume 200 mm3) were treated with Tamoxifen (citrate) by oral gavage. Tumor volume (length×width2/2) and body weight were monitored twice weekly. Following a significant anti-tumor response in which tumor volume remained static, evident tumor growth was first observed at approximately 100 days of treatment. At 120 days of treatment, tamoxifen dose was increased. Rapidly growing tumors were deemed tamoxifen resistant and selected for in vivo passage into new host animals. Tumor Fragments (−100 mm3/animal) from the tamoxifen resistant tumors were subcutaneously implanted into the right flank of female nu/nu mice (with 17-0 Estradiol pellets (0.72 mg; 60 day slow release)). Passaged tumors were maintained under constant Tamoxifen selection, and Tumor volume (length×width2/2) was monitored weekly. When tumor volume reached ˜150-250 mm3, animals were randomized into treatment groups (mean tumor volume 200 mm3) and tamoxifen treatment was terminated (except for a tamoxifen control arm) Animals were treated with Vehicle or Compound daily for 4 weeks. Tumor volume and body weight were monitored twice weekly for the duration of the study. At the conclusion of the treatment period; plasma and tumor samples were taken for pharmacokinetic and pharmacodynamic analyses, respectively.
  • Example 51 Ovarian Cancer Model; Xenograft Assay (BG-1)
  • Time release pellets (0.72 mg 17-0 Estradiol/60 days) were subcutaneously implanted into female nu/nu mice. BG-1 cells were grown in DMEM Ham's F-12 50/50 containing 10% FBS, 10 mM Sodium Pyruvate, 10 mM Non-Essential Amino Acids at 5% CO2, 37° C. Cells were spun down and re-suspended in 50% DMEM Ham's F-12 (serum free) and 50% Matrigel at 5×107 cells/mL. BG-1 cells were subcutaneously injected (100 μL/animal) on the right flank 2-3 days post pellet implantation. Tumor volume (length×width2/2) was monitored bi-weekly. When tumors reached an average volume of ˜250 mm3 animals were randomized and treatment was started. Animals were treated with Vehicle or Compound daily for 4 weeks. Tumor volume and body weight were monitored bi-weekly throughout the study. At the conclusion of the treatment period; plasma and tumor samples were taken for pharmacokinetic and pharmacodynamic analyses, respectively.
  • Example 52 Immature Uterine Wet Weight-Antagonist Mode
  • Female immature CD-IGS rats (21 days old upon arrival) were treated for three days Animals were dosed daily for three days. Vehicle or test compound was administered orally by gavage followed 15 minutes later by an oral dose of 0.1 mg/kg Ethynyl Estradiol. On the fourth day 24 hours after dose, plasma was collected for pharmacokinetic analysis. Immediately following plasma collection, the animals were euthanized and the uterus was removed and weighed.
  • Example 53 Immature Uterine Wet Weight-Agonist Mode
  • Female immature CD-IGS rats (21 days old upon arrival) were treated for three days. Animals were dosed daily for three days. Vehicle or test compound was administered orally by gavage followed 15 minutes later by a second oral dose of vehicle. On the fourth day 24 hours after dose, plasma was collected for pharmacokinetic analysis Immediately following plasma collection, the animals were euthanized and the uterus was removed and weighed.
  • Example 54 Breast Cancer Clinical Trial
  • Purpose: The purpose of this study is to assess the efficacy of treatment of metastatic breast cancer with a compound of Formula (I), or a pharmaceutically acceptable salt thereof, collect information on any side effects the compound of Formula (I), or a pharmaceutically acceptable salt thereof, may cause, and evaluate the pharmacokinetic properties of the compound of Formula (I), or a pharmaceutically acceptable salt thereof.
  • Intervention: Patients are administered 1-50 mg/kg of a compound of Formula (I), or a pharmaceutically acceptable salt thereof, per day or twice a day.
  • Outcome Measures: Primary Outcome Measures: The primary outcome measure is the patient's tumor response and/or disease control to a compound of Formula (I), or a pharmaceutically acceptable salt thereof, as first- and/or second-line treatment in patients with estrogen receptor (ER) positive metastatic breast cancer.
  • Secondary Outcome Measures: The second outcome measures are (a) an evaluation of the side-effects of a compound of Formula (I), or a pharmaceutically acceptable salt thereof; (b) an evaluation of the pharmacokinetic properties of a compound of Formula (I), or a pharmaceutically acceptable salt thereof; (c) an evaluation of the proportion of patients that have complete or partial response or stable disease at defined timepoint; (d) an evaluation of the time to progression and overall survival of patients treated with a compound of Formula (I), or a pharmaceutically acceptable salt thereof; and (e) biomarkers predictive of clinical response.
  • Detailed Description: Patients will be given a compound of Formula (I), or a pharmaceutically acceptable salt thereof, orally once or twice a day. Prior to each dosing cycle of a compound of Formula (I), or a pharmaceutically acceptable salt thereof, a physical exam, blood work and assessment of any side effects will be performed. Every 12 weeks the patient's cancer will be re-evaluated with either a CT scan or MRI to determine whether the treatment is working. Participation in this study will last until disease progression or unacceptable toxicity.
  • Eligibility: Female subjects, 18 years and older.
  • Inclusion Criteria: Histologically or cytologically confirmed diagnosis of invasive breast cancer, Stage IV disease. At least one measurable target lesion as defined by RECIST that has not been previously treated with local therapy. Post-menopausal status. ER positive breast cancer. HER2-negative breast cancer. Up to one prior hormonal therapy for advanced or metastatic disease. ECOG performance status 0-1. Life expectancy>12 weeks. Adequate liver and bone marrow function: AST<2.5×ULN; Bilirubin<1.5×ULN; ANC>1,500/ul; platelet count>100,000/ul; normal PT and PTT. At least 2 weeks since prior radiation and recovered from treatment-related toxicity.
  • Exclusion Criteria: HER2-positive breast cancer. Prior chemotherapy regimen for metastatic disease. History of, or presence of brain metastases. Concurrent investigational drug treatment. Prior bone marrow or stem cell transplant. History of other malignancy within the last 5 years, not including curatively-treated carcinoma in situ of the cervix or non-melanoma skin cancer. Uncontrolled infection. Active bleeding, or history of bleeding requiring transfusion.
  • Active cardiac disease. Serious medical or psychiatric illness.
  • Example 55 Parenteral Pharmaceutical Composition
  • To prepare a parenteral pharmaceutical composition suitable for administration by injection (subcutaneous, intravenous), 100 mg of a compound of Formula (I), or a water-soluble salt of a compound of Formula (I), is dissolved in sterile water and then mixed with 10 mL of 0.9% sterile saline. The mixture is incorporated into a dosage unit form suitable for administration by injection
  • In another embodiment, the following ingredients are mixed to form an injectable formulation: 1.2 g of a compound of Formula (I), or a pharmaceutically acceptable salt thereof, 2.0 mL of sodium acetate buffer solution (0.4 M), HCl (1 N) or NaOH (1 M) (q.s. to suitable pH), water (distilled, sterile) (q.s. to 20 mL). All of the above ingredients, except water, are combined and stirred and if necessary, with slight heating if necessary. A sufficient quantity of water is then added.
  • Example 56 Oral Solution
  • To prepare a pharmaceutical composition for oral delivery, an aqueous 20% propylene glycol solution is prepared. To this is added a sufficient amount of compound of Formula (I), or a pharmaceutically acceptable salt thereof, to provide a 20 mg/mL solution.
  • Example 57 Oral Capsule
  • To prepare a pharmaceutical composition for oral delivery, 100-500 mg of a compound of Formula (I), or a pharmaceutically acceptable salt thereof, is mixed with starch. The mixture is incorporated into an oral dosage unit such as a hard gelatin capsule, which is suitable for oral administration. In another embodiment, 100-500 mg of a compound of Formula (I), or a pharmaceutically acceptable salt thereof, is placed into Size 4 capsule, or size 1 capsule (hypromellose or hard gelatin) and the capsule is closed.
  • Example 58 Oral Tablet
  • A tablet is prepared by mixing 48% by weigh of a compound of Formula (I), or a pharmaceutically acceptable salt thereof, 45% by weight of microcrystalline cellulose, 5% by weight of low-substituted hydroxypropyl cellulose, and 2% by weight of magnesium stearate. Tablets are prepared by direct compression. The total weight of the compressed tablets is maintained at 250-500 mg.
  • Example 59 Topical Gel Composition
  • To prepare a pharmaceutical topical gel composition, a compound of Formula (I), or a pharmaceutically acceptable salt thereof, is mixed with hydroxypropyl celluose, propylene glycol, isopropyl myristate and purified alcohol USP. The resulting gel mixture is then incorporated into containers, such as tubes, which are suitable for topical administration.
  • The examples and embodiments described herein are for illustrative purposes only and various modifications or changes suggested to persons skilled in the art are to be included within the spirit and purview of this application and scope of the appended claims.

Claims (16)

What is claimed is:
1. A compound of Formula (I), or a pharmaceutically acceptable salt, or N-oxide thereof:
Figure US20120071535A1-20120322-C00259
wherein,
Z is —OH or —OR10;
R2 is C1-C4alkyl, C1-C4-fluoroalkyl, C1-C4deuteroalkyl, C3-C6cycloalkyl, or —C1-C4alkylene-W;
W is hydroxy, halogen, CN, C1-C4alkyl, C1-C4haloalkyl, C1-C4alkoxy, C1-C4haloalkoxy, or C3-C6cycloalkyl;
each R3 is independently halogen, C1-C4alkyl, or C1-C4-fluoroalkyl;
each R4 is independently halogen, —CN, —OR9, —S(═O)2R10, C1-C4alkyl, C1-C4-fluoroalkyl, or C1-C4heteroalkyl;
each R5 is independently halogen, —CN, —OR9, —S(═O)2R10, C1-C4alkyl, C1-C4-fluoroalkyl, or Cr C4heteroalkyl;
R6 is H, C1-C4alkyl, or halogen;
R7 is H, C1-C4alkyl, or halogen;
R9 is H, C1-C6alkyl, C1-C6-fluoroalkyl, or C3-C6cycloalkyl;
R10 is C1-C6alkyl;
m is 0, 1, or 2;
n is 0, 1, 2, 3, or 4; and
p is 0, 1, or 2.
2. The compound of claim 1, or a pharmaceutically acceptable salt, or N-oxide thereof, wherein:
Z is —OH;
R6 is H, —CH3, F, or Cl;
R7 is H, —CH3, F, or Cl;
each R3 is independently halogen, C1-C4alkyl, or C1-C4-fluoroalkyl;
each R4 is independently halogen, —CN, —OH, —OR9, —S(═O)2R10, C1-C4alkyl, C1-C4-fluoroalkyl, Cr C4heteroalkyl, C1-C4-fluoroalkoxy, or C1-C4alkoxy;
each R5 is independently halogen, C1-C4alkyl, or C1-C4-fluoroalkyl;
m is 0 or 1;
n is 0, 1, or 2; and
p is 0 or 1.
3. The compound of claim 2, or a pharmaceutically acceptable salt, or N-oxide thereof, wherein:
R2 is C1-C4alkyl, C1-C4-fluoroalkyl, C1-C4deuteroalkyl, C3-C6cycloalkyl, or —C1-C4alkylene-W;
W is hydroxy, halogen, CN, C1-C4alkoxy, or C3-C6cycloalkyl.
4. The compound of claim 3, or a pharmaceutically acceptable salt, or N-oxide thereof, wherein:
Z is —OH;
R6 is H;
R7 is H;
m is 0;
n is 0, 1, or 2; and
p is 0.
5. The compound of claim 1, wherein the compound of Formula (I) has the structure of Formula (II), or a pharmaceutically acceptable salt, or N-oxide thereof:
Figure US20120071535A1-20120322-C00260
6. The compound of claim 5, or a pharmaceutically acceptable salt, or N-oxide thereof, wherein:
each R3 is independently F, Cl, or —CH3;
each R4 is independently halogen, —CN, —OH, —S(═O)2CH3, —S(═O)2CH2CH3, —CH3, —CH2CH3, —CF3, —CH2OH, —OCF3, —OCH3, or —OCH2CH3;
each R5 is independently F, Cl, or —CH3;
m is 0 or 1;
n is 0, 1, or 2; and
p is 0 or 1.
7. The compound of claim 6, or a pharmaceutically acceptable salt, or N-oxide thereof, wherein:
R2 is —CH3, —CH2CH3, —CH2CH2CH3, —CH(CH3)2, —CH2CH2CH2CH3, —CH2CH(CH3)2, —CH2F, —CHF2, —CF3, —CH2CF3, —CD3, —CH2CD3, —CD2CD3, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, —CH2—W, or —CH2CH2—W;
W is hydroxy, F, Cl, —CN, —OCH3, —OCH2CH3, —OCH2CH2CH3, —OCH(CH3)2, cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl.
8. The compound of claim 5, or a pharmaceutically acceptable salt, or N-oxide thereof, wherein:
R2 is —CH2CH3;
each R4 is independently F, Cl, —CN, —OH, —CH3, —CH2CH3, —CF3, —CH2OH, —OCF3, —OCH3, or —OCH2CH3;
m is 0;
n is 0, 1, or 2; and
p is 0.
9. The compound of claim 1, wherein the compound is: (E)-Ethyl 3-(4-((E)-1-(1H-indazol-5-yl)-2-phenylbut-1-en-1-yl)phenyl)acrylate; (E)-3-(4-((E)-1-(1H-Indazol-5-yl)-2-phenylbut-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-2-(4-Fluorophenyl)-1-(1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-2-(4-Chlorophenyl)-1-(1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-1-(1H-Indazol-5-yl)-2-(3-methoxyphenyl)but-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-2-(3-(Hydroxymethyl)phenyl)-1-(1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-2-(4-(Hydroxymethyl)phenyl)-1-(1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-2-(2-(Hydroxymethyl)phenyl)-1-(1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-1-(1H-Indazol-5-yl)-2-(o-tolyl)but-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-1-(1H-Indazol-5-yl)-2-(m-tolyl)but-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-1-(1H-Indazol-5-yl)-2-(p-tolyl)but-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-1-(1H-Indazol-5-yl)-2-(2-methoxyphenyl)but-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-1-(1H-Indazol-5-yl)-2-(4-methoxyphenyl)but-1-en-1-yl)phenyl)acrylic acid; ((E)-3-(4-((E)-2-(2-Chlorophenyl)-1-(1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-243-Chlorophenyl)-1-(1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-2-(2-Fluorophenyl)-1-(1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-243-Fluorophenyl)-1-(1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-2-(2-Ethylphenyl)-1-(1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-1-(1H-Indazol-5-yl)-2-(2-(trifluoromethyl)phenyl)but-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-4-Chloro-1-(1H-indazol-5-yl)-2-phenylbut-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-2-(2-Cyanophenyl)-1-(1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-2-(2,4-Difluorophenyl)-1-(1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-2-(2-Chloro-3-fluorophenyl)-1-(1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-2-Cyclopropyl-1-(1H-indazol-5-yl)-2-phenylvinyl)phenyl)acrylic acid; (E)-3-(4-((E)-2-(4-Fluoro-2-methylphenyl)-1-(1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-2-(2,6-Difluorophenyl)-1-(1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-2-(2,6-Dichlorophenyl)-1-(1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-4,4,4-Trideutero-1-(1H-indazol-5-yl)-2-phenylbut-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-2(4-Fluoro-3-methylphenyl)-1-(1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-2-(5-Fluoro-2-methylphenyl)-1-(1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-2-(2,3-Difluorophenyl)-1-(1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-2-(2,5-Difluorophenyl)-1-(1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-2-(2-Chloro-5-fluorophenyl)-1-(1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-2-(2-Chloro-6-methylphenyl)-1-(1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-1-(7-Chloro-1H-indazol-5-yl)-2-phenylbut-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-1-(4-Methyl-1H-indazol-5-yl)-2-phenylbut-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-1-(7-Methyl-1H-indazol-5-yl)-2-phenylbut-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-1-(6-Methyl-1H-indazol-5-yl)-2-phenylbut-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-1-(3-Methyl-1H-indazol-5-yl)-2-phenylbut-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-1-(3-Chloro-1H-indazol-5-yl)-2-phenylbut-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-2-(4-Chloro-2-methylphenyl)-1-(1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-1-(1H-Indazol-5-yl)-2-phenylprop-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-1-(1H-Indazol-5-yl)-2-phenylpent-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-2-(3-Cyanophenyl)-1-(1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-2-(4-Cyanophenyl)-1-(1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-4-Hydroxy-1-(1H-indazol-5-yl)-2-phenylbut-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-1-(1H-Indazol-5-yl)-4-methoxy-2-phenylbut-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((Z)-1-(1H-Indazol-5-yl)-3-methoxy-2-phenylprop-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-1-(4-Fluoro-1H-indazol-5-yl)-2-phenylbut-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-1-(6-Chloro-1H-indazol-5-yl)-2-phenylbut-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-1-(1H-Indazol-5-yl)-4-methyl-2-phenylpent-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-1-(4-Chloro-1H-indazol-5-yl)-2-phenylbut-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-2-Cyclopentyl-1-(1H-indazol-5-yl)-2-phenylvinyl)phenyl)acrylic acid; (E)-3-(4-((E)-2-Cyclohexyl-1-(1H-indazol-5-yl)-2-phenylvinyl)phenyl)acrylic acid; (E)-3-(4-((E)-1-(1H-Indazol-5-yl)-3-methyl-2-phenylbut-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-3-Cyclopropyl-1-(1H-indazol-5-yl)-2-phenylprop-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-2-(2-Chlorophenyl)-2-cyclopropyl-1-(1H-indazol-5-yl)vinyl)phenyl)acrylic acid; (E)-3-(4-((E)-1-(6-Fluoro-1H-indazol-5-yl)-2-phenylbut-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-1-(1H-Indazol-5-yl)-2-phenylhex-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-3-Cyclopentyl-1-(1H-indazol-5-yl)-2-phenylprop-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-2-(2-Chloro-4-fluorophenyl)-1-(4-fluoro-1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-1-(7-Fluoro-1H-indazol-5-yl)-2-phenylbut-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-2-(2-Chloro-4-fluorophenyl)-1-(1H-indazol-5-yl)-4-methylpent-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((Z)-3,3-Difluoro-1-(1H-indazol-5-yl)-2-phenylprop-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-2-(2-Chloro-4-fluorophenyl)-1-(7-fluoro-1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-4-Fluoro-1-(1H-indazol-5-yl)-2-phenylbut-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-4-Chloro-2-(2-chloro-4-fluorophenyl)-1-(1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylic acid; (E)-3-(44(Z)-3,3,3-Trifluoro-1-(1H-indazol-5-yl)-2-phenylprop-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-1-(4-Fluoro-1H-indazol-5-yl)-2-(4-fluoro-2-methylphenyl)but-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-2-(4-Chloro-2-methylphenyl)-1-(4-fluoro-1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-2-Cyclopropyl-1-(4-fluoro-1H-indazol-5-yl)-2-(4-fluoro-2-methylphenyl)vinyl)phenyl)acrylic acid; (E)-3-(4-((E)-2-(4-Chloro-2-methylphenyl)-2-cyclopropyl-1-(4-fluoro-1H-indazol-5-yl)vinyl)phenyl)acrylic acid; (E)-3-(4-((E)-1-(4-Chloro-1H-indazol-5-yl)-2-(2-chloro-4-fluorophenyl)but-1-en-1-yl)phenyl)acrylic acid; (E)-3-(44(Z)-2-(2-Chloro-4-fluorophenyl)-3,3-difluoro-1-(1H-indazol-5-yl)prop-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-2-Cyclopropyl-1-(4-fluoro-1H-indazol-5-yl)-2-phenylvinyl)phenyl)acrylic acid; (E)-3-(4-((E)-4-Chloro-1-(4-fluoro-1H-indazol-5-yl)-2-phenylbut-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-4-Chloro-2-(2-chloro-4-fluorophenyl)-1-(4-fluoro-1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-4-Fluoro-2-(4-fluorophenyl)-1-(1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-4-Fluoro-1-(4-fluoro-1H-indazol-5-yl)-2-phenylbut-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-1-(1H-Indazol-5-yl)-5-methoxy-2-phenylpent-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-1-(1H-Indazol-5-yl)-6-methoxy-2-phenylhex-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-2-(2-Chloro-4-fluorophenyl)-1-(1H-indazol-5-yl)-3-methylbut-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-1-(1H-Indazol-5-yl)-2-(3-(trifluoromethoxy)phenyl)but-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-2-Cyclobutyl-1-(1H-indazol-5-yl)-2-phenylvinyl)phenyl)acrylic acid; (E)-3-(4-((E)-2-(2-Chloro-4-fluorophenyl)-2-cyclobutyl-1-(1H-indazol-5-yl)vinyl)phenyl)acrylic acid; (E)-3-(4-((E)-1-(3-Fluoro-1H-indazol-5-yl)-2-phenylbut-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-2-Cyclobutyl-1-(3-fluoro-1H-indazol-5-yl)-2-phenylvinyl)phenyl)acrylic acid; (E)-3-(4-((E)-2-(2-Chloro-4-fluorophenyl)-1-(3-fluoro-1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-2-(2-Chloro-4-fluorophenyl)-2-cyclobutyl-1-(3-fluoro-1H-indazol-5-yl)vinyl)phenyl)acrylic acid; (E)-Ethyl 3-(4-((E)-2-(4-fluorophenyl)-1-(1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylate; (E)-Ethyl 3-(4-((E)-1-(1H-Indazol-5-yl)-2-(2-methoxyphenyl)but-1-en-1-yl)phenyl)acrylate; (E)-Ethyl 3-(4-((E)-1-(1H-Indazol-5-yl)-2-(4-methoxyphenyl)but-1-en-1-yl)phenyl)acrylate; (E)-3-(4-((E)-2-(3-Hydroxyphenyl)-1-(1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-2-(2-Hydroxyphenyl)-1-(1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-2-(4-Hydroxyphenyl)-1-(1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-2-(3-Butoxyphenyl)-1-(1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-2-(4-Butoxyphenyl)-1-(1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-1-(1H-Indazol-5-yl)-2-(2-(methylsulfonyl)phenyl)but-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-1-(1H-Indazol-5-yl)-2-phenylbut-1-en-1-yl)phenyl)-2-methylacrylic acid; (E)-3-(4-((Z)-1-(1H-Indazol-5-yl)-2-phenylbut-1-en-1-yl)-3-methylphenyl)acrylic acid; (E)-3-(4-((E)-1-(1H-Indazol-5-yl)-2-phenylbut-1-en-1-yl)-2-methylphenyl)acrylic acid; (E)-3-(4-((Z)-1-(1H-Indazol-5-yl)-2-phenylbut-1-en-1-yl)-2-chlorophenyl)acrylic acid; (Z)-3-(4-((E)-1-(1H-Indazol-5-yl)-2-phenylbut-1-en-1-yl)phenyl)-2-fluoroacrylic acid; (Z)-3-(4-((E)-1-(1H-Indazol-5-yl)-2-phenylbut-1-en-1-yl)phenyl)-2-chloroacrylic acid; (E)-3-(4-((Z)-1-(1H-Indazol-5-yl)-2-phenylbut-1-en-1-yl)-3-fluorophenyl)acrylic acid; (E)-3-(4-((Z)-1-(1H-Indazol-5-yl)-2-phenylbut-1-en-1-yl)-2-fluorophenyl)acrylic acid; (E)-3-(4-((Z)-1-(1H-Indazol-5-yl)-2-phenylbut-1-en-1-yl)-2-(trifluoromethyl)phenyl)acrylic acid; (E)-3-(4-((Z)-1-(1H-Indazol-5-yl)-2-phenylbut-1-en-1-yl)-3-methoxyphenyl)acrylic acid; (E)-3-(4-((Z)-1-(1H-Indazol-5-yl)-2-phenylbut-1-en-1-yl)-2-methoxyphenyl)acrylic acid; (E)-Ethyl 3-(4-((E)-2-(2-chloro-4-fluorophenyl)-1-(1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylate hydrochloride; (E)-3-(4-((E)-2-(2-Chloro-4-fluorophenyl)-1-(1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylic acid; (E)-Ethyl 3-(4-((E)-2-(2,4-dichlorophenyl)-1-(1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylate; (E)-3-(4-((E)-2-(2,4-Dichlorophenyl)-1-(1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-2-(4-Chloro-2-(trifluoromethyl)phenyl)-1-(1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-2-(2-Chloro-4-fluorophenyl)-2-cyclopropyl-1-(1H-indazol-5-yl)vinyl)phenyl)acrylic acid; (E)-3-(4-((E)-2-(4-Fluoro-2-(trifluoromethyl)phenyl)-1-(1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-(1-(4-Fluoro-1H-indazol-5-yl)-2-(4-fluoro-2-(trifluoromethyl)phenyl)butyl)phenyl)acrylic acid; (E)-3-(4-((E)-2-(2,4-Dichlorophenyl)-1-(4-fluoro-1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-2-(4-Chloro-2-(trifluoromethyl)phenyl)-1-(4-fluoro-1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-2-(2-Chloro-4-fluorophenyl)-4-fluoro-1-(1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-2-(2-Chloro-4-methoxyphenyl)-1-(1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-2-(2,4-Dichlorophenyl)-4-fluoro-1-(4-fluoro-1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-2-Cyclopropyl-2-(2,4-dichlorophenyl)-1-(4-fluoro-1H-indazol-5-yl)vinyl)phenyl)acrylic acid; (E)-3-(4-((E)-2-(2-Chloro-4-fluorophenyl)-2-cyclopropyl-1-(4-fluoro-1H-indazol-5-yl)vinyl)phenyl)acrylic acid; (E)-3-(4-((E)-2-Cyclopropyl-2-(2,4-dichlorophenyl)-1-(1H-indazol-5-yl)vinyl)phenyl)acrylic acid; (E)-3-(4-((E)-2-(4-Chloro-2-methylphenyl)-2-cyclopropyl-1-(1H-indazol-5-yl)vinyl)phenyl)acrylic acid; (E)-3-(4-((E)-1-(1H-Indazol-5-yl)-2-(2-methyl-5-(methylsulfonyl)phenyl)but-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-1-(1H-Indazol-5-yl)-2-(4-methoxy-2-methylphenyl)but-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-2-(2-Fluoro-4-methoxyphenyl)-1-(1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-2-(2-Chloro-5-methoxyphenyl)-1-(1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-2-(2-Fluoro-4-(methylsulfonyl)phenyl)-1-(1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-2-(2,4-Dichlorophenyl)-3,3,4,4,4-pentadeutero-1-(1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-1-(1H-Indazol-5-yl)-2-(3-(methylsulfonyl)phenyl)but-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-2-(2,4-Dichlorophenyl)-1-(7-fluoro-1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-2-(2-Chloro-3-methoxyphenyl)-1-(1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-2-(2-Chloro-4-fluorophenyl)-3,3,4,4,4-pentadeutero-1-(1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-2-(4-Chloro-2-cyanophenyl)-1-(1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-2-(2-Cyano-4-fluorophenyl)-1-(1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-2-(2-Cyano-4-(trifluoromethyl)phenyl)-1-(1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-2-(2-Chloro-4-cyanophenyl)-1-(1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-2-(3-Cyano-2-methylphenyl)-1-(1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-2-(4-Cyano-2-methylphenyl)-1-(1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-2-(5-Cyano-2-methylphenyl)-1-(1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-2-(2-Cyano-4-methoxyphenyl)-1-(1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-2-(2-Chlorophenyl)-1-(1-methyl-1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-2-Cyclobutyl-1-(1-methyl-1H-indazol-5-yl)-2-phenylvinyl)phenyl)acrylic acid; (E)-3-(4-((E)-2-(2-Chloro-4-fluorophenyl)-1-(1-methyl-1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylic acid; (E)-3-(4-((E)-2-(2-Chloro-4-fluorophenyl)-1-(1-(difluoromethyl)-1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylic acid; or a pharmaceutically acceptable salt, or N-oxide thereof.
10. A method of treating cancer in a mammal comprising administering a therapeutically effective amount of a compound according to claim 1, or a pharmaceutically acceptable salt, or N-oxide thereof, to the mammal in need thereof.
11. The method of claim 10, wherein the cancer is amenable to treatment with an estrogen receptor modulator.
12. The method of claim 10, wherein the cancer is breast cancer, ovarian cancer, endometrial cancer, prostate cancer, or uterine cancer.
13. A method of treating bone cancer, breast cancer, colorectal cancer, endometrial cancer, prostate cancer, ovarian cancer, uterine cancer, cervical cancer, lung cancer, leiomyoma, uterine leiomyoma, alcoholism, migraine, aortic aneurysm, susceptibility to myocardial infarction, aortic valve sclerosis, cardiovascular disease, coronary artery disease, hypertension, deep vein thrombosis, Graves' Disease, arthritis, multiple sclerosis, cirrhosis, hepatitis B, chronic liver disease, bone density, cholestasis, hypospadias, obesity, osteoarthritis, osteopenia, osteoporosis, Alzheimer's disease, Parkinson's disease, migraine, vertigo, anorexia nervosa, attention deficit hyperactivity disorder (ADHD), dementia, major depressive disorder, psychosis, age of menarche, endometriosis, or infertility in a mammal comprising administering a therapeutically effective amount of a compound according to claim 1, or a pharmaceutically acceptable salt, or N-oxide thereof, to the mammal in need thereof.
14. A pharmaceutical composition comprising a compound of claim 1, or a pharmaceutically acceptable salt, or N-oxide thereof.
15. The pharmaceutical composition of claim 14, wherein the pharmaceutical composition is formulated for intravenous injection, subcutaneous injection, oral administration, or topical administration.
16. The pharmaceutical composition of claim 14, wherein the pharmaceutical composition is a tablet, a pill, a capsule, a liquid, a suspension, a gel, a dispersion, a solution, an emulsion, an ointment, or a lotion.
US13/234,035 2010-09-16 2011-09-15 Estrogen receptor modulators and uses thereof Active US8299112B2 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US13/234,035 US8299112B2 (en) 2010-09-16 2011-09-15 Estrogen receptor modulators and uses thereof
US13/613,467 US8455534B2 (en) 2010-09-16 2012-09-13 Estrogen receptor modulators and uses thereof

Applications Claiming Priority (7)

Application Number Priority Date Filing Date Title
US38365910P 2010-09-16 2010-09-16
US41072710P 2010-11-05 2010-11-05
US201161446967P 2011-02-25 2011-02-25
GB1104288 2011-03-15
GB1104288.4A GB2483736B (en) 2010-09-16 2011-03-15 Estrogen receptor modulators and uses thereof
GB1104288.4 2011-03-15
US13/234,035 US8299112B2 (en) 2010-09-16 2011-09-15 Estrogen receptor modulators and uses thereof

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US13/613,467 Division US8455534B2 (en) 2010-09-16 2012-09-13 Estrogen receptor modulators and uses thereof

Publications (2)

Publication Number Publication Date
US20120071535A1 true US20120071535A1 (en) 2012-03-22
US8299112B2 US8299112B2 (en) 2012-10-30

Family

ID=46599311

Family Applications (4)

Application Number Title Priority Date Filing Date
US13/234,035 Active US8299112B2 (en) 2010-09-16 2011-09-15 Estrogen receptor modulators and uses thereof
US13/821,190 Active 2033-02-23 US9399646B2 (en) 2010-09-16 2011-09-15 Estrogen receptor modulators and uses thereof
US13/613,467 Active US8455534B2 (en) 2010-09-16 2012-09-13 Estrogen receptor modulators and uses thereof
US15/186,246 Active US9868704B2 (en) 2010-09-16 2016-06-17 Estrogen receptor modulators and uses thereof

Family Applications After (3)

Application Number Title Priority Date Filing Date
US13/821,190 Active 2033-02-23 US9399646B2 (en) 2010-09-16 2011-09-15 Estrogen receptor modulators and uses thereof
US13/613,467 Active US8455534B2 (en) 2010-09-16 2012-09-13 Estrogen receptor modulators and uses thereof
US15/186,246 Active US9868704B2 (en) 2010-09-16 2016-06-17 Estrogen receptor modulators and uses thereof

Country Status (30)

Country Link
US (4) US8299112B2 (en)
EP (2) EP2735562B2 (en)
JP (2) JP5903435B2 (en)
KR (1) KR20130136986A (en)
CN (1) CN103189361B (en)
AU (1) AU2011301952B2 (en)
BR (1) BR112013006395A2 (en)
CA (1) CA2810094A1 (en)
CL (1) CL2013000722A1 (en)
CO (1) CO6680708A2 (en)
CR (1) CR20130168A (en)
DK (1) DK2735562T3 (en)
EA (1) EA023947B1 (en)
ES (1) ES2535155T3 (en)
GB (1) GB2483736B (en)
HK (1) HK1192886A1 (en)
HR (1) HRP20150433T1 (en)
HU (1) HUE025746T2 (en)
IL (1) IL224976A (en)
MX (1) MX2013002975A (en)
NZ (1) NZ607795A (en)
PE (1) PE20140191A1 (en)
PL (1) PL2735562T3 (en)
PT (1) PT2735562E (en)
RS (1) RS54026B1 (en)
SG (1) SG188551A1 (en)
SI (1) SI2735562T1 (en)
TW (1) TWI439268B (en)
WO (2) WO2012037411A2 (en)
ZA (1) ZA201301623B (en)

Cited By (28)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9018244B2 (en) 2011-12-16 2015-04-28 Olema Pharmaceuticals, Inc. Benzopyran compounds, compositions and uses thereof
WO2015082990A1 (en) * 2013-12-06 2015-06-11 Seragon Pharmaceuticals, Inc. Estrogen receptor modulator for the treatment of locally advanced or metastatic estrogen receptor positive breast cancer
US20160039770A1 (en) * 2014-08-11 2016-02-11 Genentech, Inc. Crystalline forms of an estrogen receptor modulator
US9399646B2 (en) 2010-09-16 2016-07-26 Genentech, Inc. Estrogen receptor modulators and uses thereof
US9475798B2 (en) 2013-06-19 2016-10-25 Genentech, Inc. Azetidine estrogen receptor modulators and uses thereof
US9855270B2 (en) 2014-03-13 2018-01-02 Genentech, Inc. Methods and compositions for modulating estrogen receptor mutants
US9873684B2 (en) 2013-06-19 2018-01-23 Genentech, Inc. Estrogen receptor modulators and uses thereof
US9975856B2 (en) 2015-10-07 2018-05-22 Genentech, Inc. Process for the preparation of (E)-3-(4-((E)-2-(2-chloro-4-fluorophenyl)-1-(1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylic acid
US10053451B2 (en) 2015-05-26 2018-08-21 Genentech, Inc. Heterocyclic estrogen receptor modulators and uses thereof
US10118910B2 (en) 2015-12-09 2018-11-06 The Board Of Trustees Of The University Of Illinois Benzothiophene-based selective estrogen receptor downregulators
WO2019099838A1 (en) 2017-11-16 2019-05-23 Novartis Ag Combination therapies
CN110267940A (en) * 2016-11-24 2019-09-20 卫材 R&D 管理有限公司 The purposes of quaternary olefin(e) compound and they
WO2020012337A1 (en) 2018-07-10 2020-01-16 Novartis Ag 3-(5-amino-1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives and their use in the treatment of i karos family zinc finger 2 (ikzf2)-dependent diseases
WO2020128972A1 (en) 2018-12-20 2020-06-25 Novartis Ag Dosing regimen and pharmaceutical combination comprising 3-(1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives
WO2020165833A1 (en) 2019-02-15 2020-08-20 Novartis Ag 3-(1-oxo-5-(piperidin-4-yl)isoindolin-2-yl)piperidine-2,6-dione derivatives and uses thereof
WO2020165834A1 (en) 2019-02-15 2020-08-20 Novartis Ag Substituted 3-(1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives and uses thereof
WO2020236817A2 (en) 2019-05-20 2020-11-26 Novartis Ag Mcl-1 inhibitor antibody-drug conjugates and methods of use
WO2021123996A1 (en) 2019-12-20 2021-06-24 Novartis Ag Uses of anti-tgf-beta antibodies and checkpoint inhibitors for the treatment of proliferative diseases
WO2021260528A1 (en) 2020-06-23 2021-12-30 Novartis Ag Dosing regimen comprising 3-(1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives
WO2022029573A1 (en) 2020-08-03 2022-02-10 Novartis Ag Heteroaryl substituted 3-(1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives and uses thereof
WO2022112942A2 (en) 2020-11-24 2022-06-02 Novartis Ag Anti-cd48 antibodies, antibody drug conjugates, and uses thereof
WO2022115451A1 (en) 2020-11-24 2022-06-02 Novartis Ag Mcl-1 inhibitor antibody-drug conjugates and methods of use
US11364222B2 (en) 2017-01-06 2022-06-21 G1 Therapeutics, Inc. Combination therapy for treatment of cancer
WO2022169780A1 (en) 2021-02-02 2022-08-11 Les Laboratoires Servier Selective bcl-xl protac compounds and methods of use
WO2022215011A1 (en) 2021-04-07 2022-10-13 Novartis Ag USES OF ANTI-TGFβ ANTIBODIES AND OTHER THERAPEUTIC AGENTS FOR THE TREATMENT OF PROLIFERATIVE DISEASES
WO2022243846A1 (en) 2021-05-18 2022-11-24 Novartis Ag Combination therapies
WO2023225336A1 (en) 2022-05-20 2023-11-23 Novartis Ag Met bcl-xl inhibitor antibody-drug conjugates and methods of use thereof
WO2023225320A1 (en) 2022-05-20 2023-11-23 Novartis Ag Epha2 bcl-xl inhibitor antibody-drug conjugates and methods of use thereof

Families Citing this family (56)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
SG185637A1 (en) * 2010-06-10 2012-12-28 Aragon Pharmaceuticals Inc Estrogen receptor modulators and uses thereof
WO2013056178A2 (en) 2011-10-14 2013-04-18 Foundation Medicine, Inc. Novel estrogen receptor mutations and uses thereof
US9187460B2 (en) 2011-12-14 2015-11-17 Seragon Pharmaceuticals, Inc. Estrogen receptor modulators and uses thereof
KR20140138296A (en) * 2012-03-20 2014-12-03 세라곤 파마슈티컬스, 인크. Estrogen receptor modulators and uses thereof
MX2015011132A (en) * 2013-03-14 2015-11-30 Seragon Pharmaceuticals Inc Polycyclic estrogen receptor modulators and uses thereof.
AR097617A1 (en) 2013-09-13 2016-04-06 Actelion Pharmaceuticals Ltd ANTIBACTERIAL DERIVATIVES OF 2H-INDAZOL
MA39189A1 (en) 2013-12-19 2017-06-30 Actelion Pharmaceuticals Ltd Derivatives of 1h-indazole and 1h-indole antibacterials
RU2016137122A (en) * 2014-03-13 2018-04-18 Ф. Хоффманн-Ля Рош Аг THERAPEUTIC COMBINATIONS WITH MODULATORS OF ESTROGEN RECEPTORS
WO2016024350A1 (en) 2014-08-13 2016-02-18 株式会社エス・ディー・エス バイオテック Condensed 11-membered ring compounds and agricultural and horticultural fungicide containing same
JP6359940B2 (en) * 2014-10-14 2018-07-18 国立大学法人 名古屋工業大学 Difluoromethylacetylene compounds and process for producing the same
DK3302471T3 (en) * 2015-05-29 2021-11-15 Eisai R&D Man Co Ltd TETRA-SUBSTITUTED ALKENE COMPOUNDS AND THEIR USE
CN105061316B (en) * 2015-07-17 2017-12-22 苏州大学 Thick cyclics, preparation method and purposes
KR101771794B1 (en) 2015-08-04 2017-08-25 경북대학교 산학협력단 Triazolo-arylacrylonitrile derivatives and use thereof
CN106518768B (en) * 2015-09-15 2021-07-02 江苏恒瑞医药股份有限公司 Acrylic acid derivative, preparation method and medical application thereof
UA122346C2 (en) 2015-10-01 2020-10-26 Олема Фармасьютикалз, Інк. TETRAHYDRO-1H-PYRIDO[3,4-b]INDOLE ANTI-ESTROGENIC DRUGS
KR20180083863A (en) 2015-10-16 2018-07-23 에자이 알앤드디 매니지먼트 가부시키가이샤 EP4 antagonist
US10519148B2 (en) 2015-11-12 2019-12-31 Zhejiang Hisun Pharmaceutical Co., Ltd. Acrylic acid derivative, preparation method and use in medicine thereof
WO2017138069A1 (en) 2016-02-08 2017-08-17 株式会社エス・ディー・エス バイオテック Germicidal composition
CN108602765B (en) 2016-02-08 2022-05-03 高文有限公司 Method for producing 1, 2-benzenedimethanol compound
DK3416962T3 (en) 2016-02-15 2021-07-26 Sanofi Sa 6,7-DIHYDRO-5H-BENZO [7] ANNULENE DERIVATIVES AS ESTROGEN RECEPTOR MODULATORS
RU2722441C2 (en) * 2016-03-25 2020-06-01 Лосинь Фармасьютикал (Шанхай) Ко., Лтд. Substituted indole compounds as reducing regulators of estrogen receptors
WO2017197046A1 (en) 2016-05-10 2017-11-16 C4 Therapeutics, Inc. C3-carbon linked glutarimide degronimers for target protein degradation
CN109562107A (en) 2016-05-10 2019-04-02 C4医药公司 Heterocycle degron body for target protein degradation
CN109562113A (en) 2016-05-10 2019-04-02 C4医药公司 Loop coil degron body for target protein degradation
BR112018077136A2 (en) 2016-07-01 2019-04-30 G1 Therapeutics, Inc. compound, and methods for treating a disorder associated with abnormal cell proliferation and for reducing the effect of chemotherapy on healthy cells.
CN107973780B (en) * 2016-10-21 2020-10-09 四川科伦博泰生物医药股份有限公司 Substituted olefin compound and preparation method and application thereof
WO2018081168A2 (en) 2016-10-24 2018-05-03 The Board Of Trustees Of The University Of Illinois Benzothiophene-based selective mixed estrogen receptor downregulators
CN106632144B (en) * 2016-10-31 2019-02-26 浙江省医学科学院 A kind of cyclopropanes compound and its preparation method and application
SG11201903908PA (en) * 2016-11-17 2019-05-30 Sanofi Sa Novel substituted n-(3-fluoropropyl)-pyrrolidine compounds, processes for their preparation and therapeutic uses thereof
MA46892A (en) * 2016-11-24 2021-04-14 Eisai R&D Man Co Ltd TETRASUBSTITUTED ALKENE COMPOUNDS AND THEIR USE FOR THE TREATMENT OF BREAST CANCER
JP6346368B1 (en) * 2016-11-28 2018-06-20 エーザイ・アール・アンド・ディー・マネジメント株式会社 Salts and crystals of indazole derivatives
TW201835064A (en) 2017-02-10 2018-10-01 美商G1治療公司 Benzothiophene estrogen receptor modulators
WO2018170447A1 (en) * 2017-03-16 2018-09-20 Eisai R&D Management Co., Ltd. Combination therapies for the treatment of breast cancer
CA3054976A1 (en) 2017-03-16 2018-09-20 Jiangsu Hengrui Medicine Co., Ltd. Heteroaryl[4,3-c]pyrimidine-5-amine derivative, preparation method therefor, and medical uses thereof
NZ759691A (en) 2017-06-29 2023-05-26 G1 Therapeutics Inc Morphic forms of git38 and methods of manufacture thereof
EP3434272A1 (en) 2017-07-25 2019-01-30 Sanofi Combination comprising palbociclib and 6-(2,4-dichlorophenyl)-5-[4-[(3s)-1-(3-fluoropropyl)pyrrolidin-3-yl]oxyphenyl]-8,9-dihydro-7h-benzo[7]annulene-2-carboxylic acid
US11034653B2 (en) 2017-09-25 2021-06-15 Luoxin Pharmaceutical (Shanghai) Co., Ltd. Crystal form of estrogen receptor inhibitor and preparation method therefor
EP3773576A4 (en) 2018-03-26 2021-12-29 C4 Therapeutics, Inc. Cereblon binders for the degradation of ikaros
CN108558838B (en) * 2018-04-20 2021-04-20 苏州凌科特新材料有限公司 Production method of estrogen receptor modulator intermediate
CN108503648B (en) * 2018-04-27 2020-04-14 安徽医科大学 Styryl pyrazolopyrimidine compound, pharmaceutical composition, preparation method and application
WO2019225552A1 (en) * 2018-05-22 2019-11-28 Eisai R&D Management Co., Ltd. Salts of indazole derivative and crystals thereof
US10487057B1 (en) * 2018-07-05 2019-11-26 Landos Biopharma, Inc. NLRX1 ligands
WO2020037203A2 (en) 2018-08-17 2020-02-20 Genentech, Inc. Diagnostic and therapeutic methods for the treatment of breast cancer
WO2020051235A1 (en) 2018-09-04 2020-03-12 C4 Therapeutics, Inc. Compounds for the degradation of brd9 or mth1
SG11202102059YA (en) 2018-09-07 2021-03-30 Sanofi Sa Salts of methyl 6-(2,4-dichlorophenyl)-5-[4-[(3s)-1-(3-fluoropropyl)pyrrolidin-3-yl]oxyphenyl]-8,9-dihydro-7h-benzo[7]annulene-2-carboxylate and preparation process thereof
US20220017463A1 (en) * 2018-11-28 2022-01-20 Luoxin Pharmaceutical (Shanghai) Co., Ltd. Salt Form of Estrogen Receptor Downregulator, Crystalline Form Thereof, and Preparation Method Therefor
JP2022513942A (en) * 2018-12-17 2022-02-09 チア タイ ティエンチン ファーマシューティカル グループ カンパニー リミテッド Estrogen receptor antagonist
WO2020132561A1 (en) 2018-12-20 2020-06-25 C4 Therapeutics, Inc. Targeted protein degradation
EP3978493A4 (en) 2019-05-24 2023-02-01 Zhejiang Hisun Pharmaceutical Co., Ltd. Crystal forms of acrylic acid derivatives, preparation method therefor and use thereof
CN110452177A (en) * 2019-09-02 2019-11-15 南通大学 A kind of synthetic method of the fluoro- 1H- indazole of the bromo- 4- of 5-
WO2022029657A1 (en) * 2020-08-04 2022-02-10 Novmetahealth Co., Ltd. Method for treatment of cytokine release syndrome
WO2022109395A2 (en) 2020-11-23 2022-05-27 Sanofi Panel of er regulated genes for use in monitoring endocrine therapy in breast cancer
CN117440950A (en) * 2021-06-27 2024-01-23 北京盛诺基医药科技股份有限公司 ER alpha receptor covalent binding antagonist
CN115960082A (en) * 2021-10-13 2023-04-14 长春金赛药业有限责任公司 Tetra-substituted olefin compound, preparation method and medical application thereof
KR20230146812A (en) * 2022-04-13 2023-10-20 재단법인 대구경북첨단의료산업진흥재단 Composition for treating inflammatory bowel disease comprising an arylethene derivative as an active ingredient
WO2024030968A1 (en) 2022-08-03 2024-02-08 Brystol-Myers Squibb Company Compounds for modulating ret protein

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008057857A1 (en) * 2006-11-01 2008-05-15 Bristol-Myers Squibb Company MODULATORS OF GLUCOCORTICOID RECEPTOR, AP-1, AND/OR NF-&kappav;B ACTIVITY AND USE THEREOF

Family Cites Families (34)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US410727A (en) 1889-09-10 beigos
US383659A (en) 1888-05-29 Armature for dynamos
FR1390978A (en) * 1963-11-20 1965-03-05 Ind Chimique Sa Process for preparing alpha- (thienyl-2) 4-hydroxystilbene and some derivatives and products obtained
EP0002097B1 (en) * 1977-08-22 1981-08-05 Imperial Chemical Industries Plc Triphenylalkene derivatives, process for their preparation and pharmaceutical compositions containing them
US4229447A (en) 1979-06-04 1980-10-21 American Home Products Corporation Intraoral methods of using benzodiazepines
US4596795A (en) 1984-04-25 1986-06-24 The United States Of America As Represented By The Secretary, Dept. Of Health & Human Services Administration of sex hormones in the form of hydrophilic cyclodextrin derivatives
US4755386A (en) 1986-01-22 1988-07-05 Schering Corporation Buccal formulation
US5739136A (en) 1989-10-17 1998-04-14 Ellinwood, Jr.; Everett H. Intraoral dosing method of administering medicaments
JP3409366B2 (en) 1993-06-18 2003-05-26 東洋インキ製造株式会社 Organic electroluminescence device
US5681835A (en) 1994-04-25 1997-10-28 Glaxo Wellcome Inc. Non-steroidal ligands for the estrogen receptor
DE19807760A1 (en) 1997-02-27 1998-09-03 Fuji Electric Co Ltd Electrophotographic photoconductor with stabilising electron acceptor, useful in printer or copier
CZ300929B6 (en) 1997-08-15 2009-09-16 Duke University Compound for use in the treatment or prevention of estrogen-stimulated cancer
DE19801597C2 (en) 1998-01-17 2001-05-03 Aventis Res & Tech Gmbh & Co Base-catalyzed synthesis of 1-aryl-4- (arylethyl) piperazines from aromatic olefins and 1-arylpiperazines
DE19801598C2 (en) 1998-01-17 2000-05-11 Aventis Res & Tech Gmbh & Co Catalytic synthesis of N-alkylated anilines from olefins and anilines
TW593256B (en) * 1999-11-16 2004-06-21 Hormos Medical Oy Ltd Triphenylalkene derivatives and their use as selective estrogen receptor modulators
GB9929988D0 (en) 1999-12-17 2000-02-09 Celltech Therapeutics Ltd Chemical compounds
US6528681B2 (en) * 2000-04-05 2003-03-04 Bristol-Meyers Squibb Pharma Company Halogenated triphenylethylene derivatives as selective estrogen receptor modulators
US6927224B2 (en) * 2001-08-11 2005-08-09 Bristol Myers Squibb Company Selective estrogen receptor modulators
AU2003248630A1 (en) 2002-06-06 2003-12-22 Smithkline Beecham Corporation Thrombopoietin mimetics
ES2307068T3 (en) * 2003-10-08 2008-11-16 Smithkline Beecham Corporation TRIFENILETILENE COMPOUNDS AS SELECTIVE MODULARS OF STROGEN RECEPTORS.
DE102004028862A1 (en) 2004-06-15 2005-12-29 Merck Patent Gmbh 3-aminoindazoles
US20070292545A1 (en) 2005-10-18 2007-12-20 Aaron Monte Anti-infective agents and methods of use
ES2336243T3 (en) * 2005-01-26 2010-04-09 Schering Corporation DERIVATIVES OF 3- (INDAZOL-5-IL) - (1,2,4) TRIAZINE AND SIMILAR COMPOSITIONS AS INHIBITORS OF PROTEIN KINASE FOR THE TREATMENT OF CANCER.
JP2009504872A (en) 2005-08-15 2009-02-05 テクニオン リサーチ アンド ディベロップメント ファウンデーション リミテッド Printable material
WO2007062151A2 (en) * 2005-11-22 2007-05-31 Smithkline Beecham Corporation Chemical compounds
JP2008156313A (en) 2006-12-26 2008-07-10 Fyuuensu:Kk Medicine for treatment and monitoring of amyloid disease
GB0704407D0 (en) 2007-03-07 2007-04-18 Glaxo Group Ltd Compounds
JP5223362B2 (en) 2008-02-07 2013-06-26 コニカミノルタビジネステクノロジーズ株式会社 Photoelectric conversion element and solar cell
US7897631B2 (en) 2008-04-21 2011-03-01 North Carolina State University Inhibition and dispersion of bacterial biofilms with imidazole-triazole derivatives
ES2548141T3 (en) 2008-11-20 2015-10-14 Glaxosmithkline Llc Chemical compounds
US20110294668A1 (en) 2008-12-08 2011-12-01 North Carolina State University Inhibition and dispersion of biofilms in plants with imidazole-triazole derivatives
WO2010107474A1 (en) 2009-03-16 2010-09-23 The Research Foundation Of State University Of New York Antiestrogens for breast cancer therapy
WO2010144959A1 (en) 2009-06-18 2010-12-23 Fibrotech Therapeutics Pty Ltd Analogues of anti-fibrotic agents
GB2483736B (en) 2010-09-16 2012-08-29 Aragon Pharmaceuticals Inc Estrogen receptor modulators and uses thereof

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008057857A1 (en) * 2006-11-01 2008-05-15 Bristol-Myers Squibb Company MODULATORS OF GLUCOCORTICOID RECEPTOR, AP-1, AND/OR NF-&kappav;B ACTIVITY AND USE THEREOF

Cited By (36)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9399646B2 (en) 2010-09-16 2016-07-26 Genentech, Inc. Estrogen receptor modulators and uses thereof
US9018244B2 (en) 2011-12-16 2015-04-28 Olema Pharmaceuticals, Inc. Benzopyran compounds, compositions and uses thereof
US9475798B2 (en) 2013-06-19 2016-10-25 Genentech, Inc. Azetidine estrogen receptor modulators and uses thereof
US9655880B2 (en) 2013-06-19 2017-05-23 Genentech, Inc. Azetidine estrogen receptor modulators and uses thereof
US9873684B2 (en) 2013-06-19 2018-01-23 Genentech, Inc. Estrogen receptor modulators and uses thereof
WO2015082990A1 (en) * 2013-12-06 2015-06-11 Seragon Pharmaceuticals, Inc. Estrogen receptor modulator for the treatment of locally advanced or metastatic estrogen receptor positive breast cancer
US9675586B2 (en) 2013-12-06 2017-06-13 Genentech, Inc. Estrogen receptor modulator for the treatment of locally advanced or metastatic estrogen receptor positive breast cancer
US10034860B2 (en) 2013-12-06 2018-07-31 Genentech, Inc. Estrogen receptor modulator for the treatment of locally advanced or metastatic estrogen receptor positive breast cancer
US9855270B2 (en) 2014-03-13 2018-01-02 Genentech, Inc. Methods and compositions for modulating estrogen receptor mutants
US20160039770A1 (en) * 2014-08-11 2016-02-11 Genentech, Inc. Crystalline forms of an estrogen receptor modulator
US9783503B2 (en) * 2014-08-11 2017-10-10 Genentech, Inc. Crystalline forms of an estrogen receptor modulator
US10053451B2 (en) 2015-05-26 2018-08-21 Genentech, Inc. Heterocyclic estrogen receptor modulators and uses thereof
US9975856B2 (en) 2015-10-07 2018-05-22 Genentech, Inc. Process for the preparation of (E)-3-(4-((E)-2-(2-chloro-4-fluorophenyl)-1-(1H-indazol-5-yl)but-1-en-1-yl)phenyl)acrylic acid
US10118910B2 (en) 2015-12-09 2018-11-06 The Board Of Trustees Of The University Of Illinois Benzothiophene-based selective estrogen receptor downregulators
US11072595B2 (en) 2015-12-09 2021-07-27 The Board of Trustees of lhe University of Illinois Benzothiophene-based selective estrogen receptor downregulator compounds
US11447461B2 (en) 2015-12-09 2022-09-20 The Board Of Trustees Of The University Of Illinois Benzothiophene-based selective estrogen receptor downregulators
US10807964B2 (en) 2015-12-09 2020-10-20 The Board Of Trustees Of The University Of Illinois Benzothiophene-based selective estrogen receptor downregulators
CN110267940A (en) * 2016-11-24 2019-09-20 卫材 R&D 管理有限公司 The purposes of quaternary olefin(e) compound and they
US11364222B2 (en) 2017-01-06 2022-06-21 G1 Therapeutics, Inc. Combination therapy for treatment of cancer
WO2019099838A1 (en) 2017-11-16 2019-05-23 Novartis Ag Combination therapies
WO2020012337A1 (en) 2018-07-10 2020-01-16 Novartis Ag 3-(5-amino-1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives and their use in the treatment of i karos family zinc finger 2 (ikzf2)-dependent diseases
WO2020128972A1 (en) 2018-12-20 2020-06-25 Novartis Ag Dosing regimen and pharmaceutical combination comprising 3-(1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives
WO2020165833A1 (en) 2019-02-15 2020-08-20 Novartis Ag 3-(1-oxo-5-(piperidin-4-yl)isoindolin-2-yl)piperidine-2,6-dione derivatives and uses thereof
WO2020165834A1 (en) 2019-02-15 2020-08-20 Novartis Ag Substituted 3-(1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives and uses thereof
WO2020236817A2 (en) 2019-05-20 2020-11-26 Novartis Ag Mcl-1 inhibitor antibody-drug conjugates and methods of use
WO2020236825A2 (en) 2019-05-20 2020-11-26 Novartis Ag Mcl-1 inhibitor antibody-drug conjugates and methods of use
WO2021123996A1 (en) 2019-12-20 2021-06-24 Novartis Ag Uses of anti-tgf-beta antibodies and checkpoint inhibitors for the treatment of proliferative diseases
WO2021260528A1 (en) 2020-06-23 2021-12-30 Novartis Ag Dosing regimen comprising 3-(1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives
WO2022029573A1 (en) 2020-08-03 2022-02-10 Novartis Ag Heteroaryl substituted 3-(1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives and uses thereof
WO2022115451A1 (en) 2020-11-24 2022-06-02 Novartis Ag Mcl-1 inhibitor antibody-drug conjugates and methods of use
WO2022112942A2 (en) 2020-11-24 2022-06-02 Novartis Ag Anti-cd48 antibodies, antibody drug conjugates, and uses thereof
WO2022169780A1 (en) 2021-02-02 2022-08-11 Les Laboratoires Servier Selective bcl-xl protac compounds and methods of use
WO2022215011A1 (en) 2021-04-07 2022-10-13 Novartis Ag USES OF ANTI-TGFβ ANTIBODIES AND OTHER THERAPEUTIC AGENTS FOR THE TREATMENT OF PROLIFERATIVE DISEASES
WO2022243846A1 (en) 2021-05-18 2022-11-24 Novartis Ag Combination therapies
WO2023225336A1 (en) 2022-05-20 2023-11-23 Novartis Ag Met bcl-xl inhibitor antibody-drug conjugates and methods of use thereof
WO2023225320A1 (en) 2022-05-20 2023-11-23 Novartis Ag Epha2 bcl-xl inhibitor antibody-drug conjugates and methods of use thereof

Also Published As

Publication number Publication date
US8299112B2 (en) 2012-10-30
HUE025746T2 (en) 2016-04-28
PL2735562T3 (en) 2015-08-31
PE20140191A1 (en) 2014-02-27
ZA201301623B (en) 2015-10-28
US20130231333A1 (en) 2013-09-05
WO2012037410A3 (en) 2012-06-28
EP2735562A1 (en) 2014-05-28
SG188551A1 (en) 2013-04-30
RS54026B1 (en) 2015-10-30
MX2013002975A (en) 2013-04-11
CN103189361B (en) 2015-09-30
WO2012037411A3 (en) 2012-07-05
JP5903435B2 (en) 2016-04-13
BR112013006395A2 (en) 2019-09-24
JP2013544761A (en) 2013-12-19
WO2012037411A9 (en) 2012-05-18
IL224976A (en) 2015-08-31
CA2810094A1 (en) 2012-03-22
NZ607795A (en) 2015-02-27
US9868704B2 (en) 2018-01-16
DK2735562T3 (en) 2015-05-11
CL2013000722A1 (en) 2013-07-26
US20130012561A1 (en) 2013-01-10
SI2735562T1 (en) 2015-07-31
US20160297767A1 (en) 2016-10-13
CN103189361A (en) 2013-07-03
TWI439268B (en) 2014-06-01
AU2011301952B2 (en) 2015-02-19
EP2735562B2 (en) 2023-11-15
WO2012037411A2 (en) 2012-03-22
CR20130168A (en) 2013-08-30
HK1192886A1 (en) 2014-09-05
TW201216961A (en) 2012-05-01
GB2483736A (en) 2012-03-21
HRP20150433T1 (en) 2015-07-31
US8455534B2 (en) 2013-06-04
CO6680708A2 (en) 2013-05-31
GB201104288D0 (en) 2011-04-27
AU2011301952A1 (en) 2013-03-21
GB2483736B (en) 2012-08-29
EA201370066A1 (en) 2014-01-30
WO2012037410A9 (en) 2012-05-10
EP2735562B1 (en) 2015-03-11
WO2012037410A2 (en) 2012-03-22
US9399646B2 (en) 2016-07-26
EP2616445A2 (en) 2013-07-24
ES2535155T3 (en) 2015-05-05
KR20130136986A (en) 2013-12-13
EA023947B1 (en) 2016-07-29
EP2616445A4 (en) 2014-01-15
PT2735562E (en) 2015-06-18
JP2016155832A (en) 2016-09-01

Similar Documents

Publication Publication Date Title
US8299112B2 (en) Estrogen receptor modulators and uses thereof
US9586952B2 (en) Polycyclic estrogen receptor modulators and uses thereof
EP3233828B1 (en) Estrogen receptor modulators and uses thereof
EP2828243B1 (en) Estrogen receptor modulators and uses thereof
EP2791133B1 (en) Fluorinated estrogen receptor modulators and uses thereof
US8703810B2 (en) Estrogen receptor modulators and uses thereof
US20130137746A1 (en) Indane estrogen receptor modulators and uses thereof
UA110219C2 (en) Estrogen receptor modulators and their use

Legal Events

Date Code Title Description
AS Assignment

Owner name: ARAGON PHARMACEUTICALS, INC., CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:SMITH, NICHOLAS D.;KAHRAMAN, MEHMET;GOVEK, STEVEN P.;AND OTHERS;SIGNING DATES FROM 20111019 TO 20111111;REEL/FRAME:027297/0499

STCF Information on status: patent grant

Free format text: PATENTED CASE

AS Assignment

Owner name: SERAGON PHARMACEUTICALS, INC., CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:ARAGON PHARMACEUTICALS, INC.;REEL/FRAME:031025/0608

Effective date: 20130816

FPAY Fee payment

Year of fee payment: 4

MAFP Maintenance fee payment

Free format text: PAYMENT OF MAINTENANCE FEE, 8TH YEAR, LARGE ENTITY (ORIGINAL EVENT CODE: M1552); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

Year of fee payment: 8

MAFP Maintenance fee payment

Free format text: PAYMENT OF MAINTENANCE FEE, 12TH YEAR, LARGE ENTITY (ORIGINAL EVENT CODE: M1553); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

Year of fee payment: 12